Pub Date : 2024-06-22DOI: 10.1016/j.jncc.2024.06.005
Hongmei Zeng , Rongshou Zheng , Kexin Sun , Maigeng Zhou , Shaoming Wang , Li Li , Ru Chen , Bingfeng Han , Meicen Liu , Jinhui Zhou , Mengyuan Xu , Lijun Wang , Peng Yin , Baohua Wang , Jinling You , Jing Wu , Wenqiang Wei , Jie He
Background
A milestone goal of the Healthy China Program (2019–2030) is to achieve 5-year cancer survival at 43.3% for all cancers combined by 2022. To assess the progress towards this target, we analyzed the updated survival for all cancers combined and 25 specific cancer types in China from 2019 to 2021.
Methods
We conducted standardized data collection and quality control for cancer registries across 32 provincial-level regions in China, and included 6,410,940 newly diagnosed cancer patients from 281 cancer registries during 2008–2019, with follow-up data on vital status available until December 2021. We estimated the age-standardized 5-year relative survival overall and by site, age group, and period of diagnosis using the International Cancer Survival Standard Weights, and quantified the survival changes to assess the progress in cancer control.
Results
In 2019–2021, the age-standardized 5-year relative survival for all cancers combined was 43.7% (95% confidence interval [CI], 43.6–43.7). The 5-year relative survival varied by cancer type, ranging from 8.5% (95% CI, 8.2–8.7) for pancreatic cancer to 92.9% (95% CI, 92.4–93.3) for thyroid cancer. Eight cancers had 5-year survival of over 60%, including cancers of the thyroid, breast, testis, bladder, prostate, kidney, uterus, and cervix. The 5-year relative survival was generally lower in males than in females. From 2008 to 2021, we observed significant survival improvements for cancers of the lung, prostate, bone, uterus, breast, cervix, nasopharynx, larynx, and bladder. The most significant improvement was in lung cancer.
Conclusions
Progress in cancer control was evident in China. This highlights the importance of a comprehensive approach to control and prevent cancer.
{"title":"Cancer survival statistics in China 2019–2021: a multicenter, population-based study","authors":"Hongmei Zeng , Rongshou Zheng , Kexin Sun , Maigeng Zhou , Shaoming Wang , Li Li , Ru Chen , Bingfeng Han , Meicen Liu , Jinhui Zhou , Mengyuan Xu , Lijun Wang , Peng Yin , Baohua Wang , Jinling You , Jing Wu , Wenqiang Wei , Jie He","doi":"10.1016/j.jncc.2024.06.005","DOIUrl":"10.1016/j.jncc.2024.06.005","url":null,"abstract":"<div><h3>Background</h3><p>A milestone goal of the Healthy China Program (2019–2030) is to achieve 5-year cancer survival at 43.3% for all cancers combined by 2022. To assess the progress towards this target, we analyzed the updated survival for all cancers combined and 25 specific cancer types in China from 2019 to 2021.</p></div><div><h3>Methods</h3><p>We conducted standardized data collection and quality control for cancer registries across 32 provincial-level regions in China, and included 6,410,940 newly diagnosed cancer patients from 281 cancer registries during 2008–2019, with follow-up data on vital status available until December 2021. We estimated the age-standardized 5-year relative survival overall and by site, age group, and period of diagnosis using the International Cancer Survival Standard Weights, and quantified the survival changes to assess the progress in cancer control.</p></div><div><h3>Results</h3><p>In 2019–2021, the age-standardized 5-year relative survival for all cancers combined was 43.7% (95% confidence interval [CI], 43.6–43.7). The 5-year relative survival varied by cancer type, ranging from 8.5% (95% CI, 8.2–8.7) for pancreatic cancer to 92.9% (95% CI, 92.4–93.3) for thyroid cancer. Eight cancers had 5-year survival of over 60%, including cancers of the thyroid, breast, testis, bladder, prostate, kidney, uterus, and cervix. The 5-year relative survival was generally lower in males than in females. From 2008 to 2021, we observed significant survival improvements for cancers of the lung, prostate, bone, uterus, breast, cervix, nasopharynx, larynx, and bladder. The most significant improvement was in lung cancer.</p></div><div><h3>Conclusions</h3><p>Progress in cancer control was evident in China. This highlights the importance of a comprehensive approach to control and prevent cancer.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 3","pages":"Pages 203-213"},"PeriodicalIF":7.6,"publicationDate":"2024-06-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000498/pdfft?md5=ba2f3e91467cd2db70ca6f11e93b38c3&pid=1-s2.0-S2667005424000498-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142048636","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-22DOI: 10.1016/j.jncc.2024.04.006
Chenyu Luo , Jiahui Luo , Yuhan Zhang , Bin Lu , Na Li , Yueyang Zhou , Shuohua Chen , Shouling Wu , Qingsong Zhang , Min Dai , Hongda Chen
Background
The incidence of early-onset colorectal cancer (EOCRC), which exhibits differential clinical, pathological, and molecular features compared to late-onset CRC (LOCRC), is rising globally. The potential differential effects of blood glucose on EOCRC compared to LOCRC have not been investigated.
Methods
This study analyzed 374,568 participants from the UK Biobank cohort and 172,809 participants from the Kailuan cohort. The linear associations between blood glucose and EOCRC/LOCRC were estimated using Cox regression models. Restricted cubic spline (RCS) analysis and non-linear Mendelian randomization (MR) analysis using a 70-SNPs genetic instrument for fasting glucose were used to explore the potential non-linear associations.
Results
Participants in the highest quintile of blood glucose had higher overall CRC risk compared to the lowest quintile (HR = 1.10 in the UK Biobank cohort, 95% CI: 1.01–1.21, P-trend = 0.012; HR = 1.23 in the Kailuan cohort, 95% CI: 1.01–1.51, P-trend = 0.036). Elevated glucose (>7.0 mmol/L) was more strongly associated with increased risk of EOCRC (HR = 1.61, 95% CI: 1.07–2.44) than with LOCRC (HR = 1.14, 95% CI: 1.02–1.27) in the UK Biobank cohort (P-heterogeneity = 0.014). Elevated glucose (>7.0 mmol/L) was associated with increased risk of LOCRC (HR = 1.25, 95% CI: 1.04–1.65) in the Kailuan cohort as well. There was no evidence for non-linear associations between blood glucose and risks of EOCRC/LOCRC.
Conclusions
This study showed a positive association between blood glucose and CRC risk in a dose-response manner, particularly for EOCRC, suggesting that tighter glucose control should be a priority for younger age groups.
{"title":"Associations between blood glucose and early- and late-onset colorectal cancer: evidence from two prospective cohorts and Mendelian randomization analyses","authors":"Chenyu Luo , Jiahui Luo , Yuhan Zhang , Bin Lu , Na Li , Yueyang Zhou , Shuohua Chen , Shouling Wu , Qingsong Zhang , Min Dai , Hongda Chen","doi":"10.1016/j.jncc.2024.04.006","DOIUrl":"10.1016/j.jncc.2024.04.006","url":null,"abstract":"<div><h3>Background</h3><p>The incidence of early-onset colorectal cancer (EOCRC), which exhibits differential clinical, pathological, and molecular features compared to late-onset CRC (LOCRC), is rising globally. The potential differential effects of blood glucose on EOCRC compared to LOCRC have not been investigated.</p></div><div><h3>Methods</h3><p>This study analyzed 374,568 participants from the UK Biobank cohort and 172,809 participants from the Kailuan cohort. The linear associations between blood glucose and EOCRC/LOCRC were estimated using Cox regression models. Restricted cubic spline (RCS) analysis and non-linear Mendelian randomization (MR) analysis using a 70-SNPs genetic instrument for fasting glucose were used to explore the potential non-linear associations.</p></div><div><h3>Results</h3><p>Participants in the highest quintile of blood glucose had higher overall CRC risk compared to the lowest quintile (HR = 1.10 in the UK Biobank cohort, 95% CI: 1.01–1.21, <em>P</em>-trend = 0.012; HR = 1.23 in the Kailuan cohort, 95% CI: 1.01–1.51, <em>P</em>-trend = 0.036). Elevated glucose (>7.0 mmol/L) was more strongly associated with increased risk of EOCRC (HR = 1.61, 95% CI: 1.07–2.44) than with LOCRC (HR = 1.14, 95% CI: 1.02–1.27) in the UK Biobank cohort (<em>P-</em>heterogeneity = 0.014). Elevated glucose (>7.0 mmol/L) was associated with increased risk of LOCRC (HR = 1.25, 95% CI: 1.04–1.65) in the Kailuan cohort as well. There was no evidence for non-linear associations between blood glucose and risks of EOCRC/LOCRC.</p></div><div><h3>Conclusions</h3><p>This study showed a positive association between blood glucose and CRC risk in a dose-response manner, particularly for EOCRC, suggesting that tighter glucose control should be a priority for younger age groups.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 3","pages":"Pages 241-248"},"PeriodicalIF":7.6,"publicationDate":"2024-06-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000486/pdfft?md5=972b697a869f545991a79c7ca79d5b77&pid=1-s2.0-S2667005424000486-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142048650","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-17DOI: 10.1016/j.jncc.2024.06.003
Aimin Jiang , Wenqiang Liu , Ying Liu , Junyi Hu , Baohua Zhu , Yu Fang , Xuenan Zhao , Le Qu , Juan Lu , Bing Liu , Lin Qi , Chen Cai , Peng Luo , Linhui Wang
Background
Emerging evidence suggests that cell deaths are involved in tumorigenesis and progression, which may be treated as a novel direction of cancers. Recently, a novel type of programmed cell death, disulfidptosis, was discovered. However, the detailed biological and clinical impact of disulfidptosis and related regulators remains largely unknown.
Methods
In this work, we first enrolled pancancer datasets and performed multi-omics analysis, including gene expression, DNA methylation, copy number variation and single nucleic variation profiles. Then we deciphered the biological implication of disulfidptosis in clear cell renal cell carcinoma (ccRCC) by machine learning. Finally, a novel agent targeting at disulfidptosis in ccRCC was identified and verified.
Results
We found that disulfidptosis regulators were dysregulated among cancers, which could be explained by aberrant DNA methylation and genomic mutation events. Disulfidptosis scores were depressed among cancers and negatively correlated with epithelial mesenchymal transition. Disulfidptosis regulators could satisfactorily stratify risk subgroups in ccRCC, and a novel subtype, DCS3, owning with disulfidptosis depression, insensitivity to immune therapy and aberrant genome instability were identified and verified. Moreover, treating DCS3 with NU1025 could significantly inhibit ccRCC malignancy.
Conclusion
This work provided a better understanding of disulfidptosis in cancers and new insights into individual management based on disulfidptosis.
{"title":"DCS, a novel classifier system based on disulfidptosis reveals tumor microenvironment heterogeneity and guides frontline therapy for clear cell renal carcinoma","authors":"Aimin Jiang , Wenqiang Liu , Ying Liu , Junyi Hu , Baohua Zhu , Yu Fang , Xuenan Zhao , Le Qu , Juan Lu , Bing Liu , Lin Qi , Chen Cai , Peng Luo , Linhui Wang","doi":"10.1016/j.jncc.2024.06.003","DOIUrl":"10.1016/j.jncc.2024.06.003","url":null,"abstract":"<div><h3>Background</h3><p>Emerging evidence suggests that cell deaths are involved in tumorigenesis and progression, which may be treated as a novel direction of cancers. Recently, a novel type of programmed cell death, disulfidptosis, was discovered. However, the detailed biological and clinical impact of disulfidptosis and related regulators remains largely unknown.</p></div><div><h3>Methods</h3><p>In this work, we first enrolled pancancer datasets and performed multi-omics analysis, including gene expression, DNA methylation, copy number variation and single nucleic variation profiles. Then we deciphered the biological implication of disulfidptosis in clear cell renal cell carcinoma (ccRCC) by machine learning. Finally, a novel agent targeting at disulfidptosis in ccRCC was identified and verified.</p></div><div><h3>Results</h3><p>We found that disulfidptosis regulators were dysregulated among cancers, which could be explained by aberrant DNA methylation and genomic mutation events. Disulfidptosis scores were depressed among cancers and negatively correlated with epithelial mesenchymal transition. Disulfidptosis regulators could satisfactorily stratify risk subgroups in ccRCC, and a novel subtype, DCS3, owning with disulfidptosis depression, insensitivity to immune therapy and aberrant genome instability were identified and verified. Moreover, treating DCS3 with NU1025 could significantly inhibit ccRCC malignancy.</p></div><div><h3>Conclusion</h3><p>This work provided a better understanding of disulfidptosis in cancers and new insights into individual management based on disulfidptosis.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 3","pages":"Pages 263-279"},"PeriodicalIF":7.6,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000462/pdfft?md5=b8b27d1b2e48460f1252ccf7d557b9d2&pid=1-s2.0-S2667005424000462-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142048651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-17DOI: 10.1016/j.jncc.2024.06.002
Xiaobin Zhu, Rajiv Trehan, Changqing Xie
Primary liver cancer is a leading cause of death worldwide. To create advanced treatments for primary liver cancer, studies have utilized models such as 2D cell culture and in vivo animal models. Recent developments in cancer organoids have created the possibility for 3D in vitro cultures that recapitulates the cancer cell structure and operation as well as the tumor microenvironment (TME). However, before organoids can be directly translated to clinical use, tissue processing and culture medium must be standardized with unified protocols to decrease variability in results. Herein, we present the wide variety of published methodologies used to derive liver cancer organoids from patient tumor tissues. Additionally, we summarize validation methodologies for organoids in terms of marker expression levels with immunohistochemistry as well as the presence of mutations and variants through RNA-sequencing. Primary liver cancer organoids have exciting applications allowing for faster drug testing at a larger scale. Primary liver cancer organoids also assisit in uncovering new mechanisms. Through the coculture of different immune cells and cancer organoids, organoids are now better able to recapitulate the liver cancer TME. In addition, it further aids in the investigation of drug development and drug resistance. Lastly, we posit that the usage of liver cancer organoids in animal models provides researchers a methodology to overcome the current limitations of culture systems.
{"title":"Primary liver cancer organoids and their application to research and therapy","authors":"Xiaobin Zhu, Rajiv Trehan, Changqing Xie","doi":"10.1016/j.jncc.2024.06.002","DOIUrl":"10.1016/j.jncc.2024.06.002","url":null,"abstract":"<div><p>Primary liver cancer is a leading cause of death worldwide. To create advanced treatments for primary liver cancer, studies have utilized models such as 2D cell culture and <em>in vivo</em> animal models. Recent developments in cancer organoids have created the possibility for 3D <em>in vitro</em> cultures that recapitulates the cancer cell structure and operation as well as the tumor microenvironment (TME)<em>.</em> However, before organoids can be directly translated to clinical use, tissue processing and culture medium must be standardized with unified protocols to decrease variability in results. Herein, we present the wide variety of published methodologies used to derive liver cancer organoids from patient tumor tissues. Additionally, we summarize validation methodologies for organoids in terms of marker expression levels with immunohistochemistry as well as the presence of mutations and variants through RNA-sequencing. Primary liver cancer organoids have exciting applications allowing for faster drug testing at a larger scale. Primary liver cancer organoids also assisit in uncovering new mechanisms. Through the coculture of different immune cells and cancer organoids, organoids are now better able to recapitulate the liver cancer TME. In addition, it further aids in the investigation of drug development and drug resistance. Lastly, we posit that the usage of liver cancer organoids in animal models provides researchers a methodology to overcome the current limitations of culture systems.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 3","pages":"Pages 195-202"},"PeriodicalIF":7.6,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000450/pdfft?md5=71d57bff340d848aa3d9e6fef9dac00e&pid=1-s2.0-S2667005424000450-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142048635","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<div><h3>Objective</h3><p>Accurate prognosis prediction is critical for individualized-therapy making of gastric cancer patients. We aimed to develop and test 6-month, 1-, 2-, 3-, 5-, and 10-year overall survival (OS) and cancer-specific survival (CSS) prediction models for gastric cancer patients following gastrectomy.</p></div><div><h3>Methods</h3><p>We derived and tested Survival Quilts, a machine learning-based model, to develop 6-month, 1-, 2-, 3-, 5-, and 10-year OS and CSS prediction models. Gastrectomy patients in the development set (<em>n</em> = 20,583) and the internal validation set (<em>n</em> = 5,106) were recruited from the Surveillance, Epidemiology, and End Results (SEER) database, while those in the external validation set (<em>n</em> = 6,352) were recruited from the China National Cancer Center Gastric Cancer (NCCGC) database. Furthermore, we selected gastrectomy patients without neoadjuvant therapy as a subgroup to train and test the prognostic models in order to keep the accuracy of tumor-node-metastasis (TNM) stage. Prognostic performances of these OS and CSS models were assessed using the Concordance Index (C-index) and area under the curve (AUC) values.</p></div><div><h3>Results</h3><p>The machine learning model had a consistently high accuracy in predicting 6-month, 1-, 2-, 3-, 5-, and 10-year OS in the SEER development set (C-index = 0.861, 0.832, 0.789, 0.766, 0.740, and 0.709; AUC = 0.784, 0.828, 0.840, 0.849, 0.869, and 0.902, respectively), SEER validation set (C-index = 0.782, 0.739, 0.712, 0.698, 0.681, and 0.660; AUC = 0.751, 0.772, 0.767, 0.762, 0.766, and 0.787, respectively), and NCCGC set (C-index = 0.691, 0.756, 0.751, 0.737, 0.722, and 0.701; AUC = 0.769, 0.788, 0.790, 0.790, 0.787, and 0.788, respectively). The model was able to predict 6-month, 1-, 2-, 3-, 5-, and 10-year CSS in the SEER development set (C-index = 0.879, 0.858, 0.820, 0.802, 0.784, and 0.774; AUC = 0.756, 0.827, 0.852, 0.863, 0.874, and 0.884, respectively) and SEER validation set (C-index = 0.790, 0.763, 0.741, 0.729, 0.718, and 0.708; AUC = 0.706, 0.758, 0.767, 0.766, 0.766, and 0.764, respectively). In multivariate analysis, the high-risk group with risk score output by 5-year OS model was proved to be a strong survival predictor both in the SEER development set (hazard ratio [HR] = 14.59, 95% confidence interval [CI]: 1.872–2.774, <em>P</em> < 0.001), SEER validation set (HR = 2.28, 95% CI: 13.089–16.293, <em>P</em> < 0.001), and NCCGC set (HR = 1.98, 95% CI: 1.617–2.437, <em>P</em> <em><</em> 0.001). We further explored the prognostic value of risk score resulted 5-year CSS model of gastrectomy patients, and found that high-risk group remained as an independent CSS factor in the SEER development set (HR = 12.81, 95% CI: 11.568–14.194, <em>P</em> < 0.001) and SEER validation set (HR = 1.61, 95% CI: 1.338–1.935, <em>P</em> < 0.001).</p></div><div><h3>Conclusion</h3><p>Survival Quilts could allow accurate prediction of 6-m
{"title":"Application of Survival Quilts for prognosis prediction of gastrectomy patients based on the Surveillance, Epidemiology, and End Results database and China National Cancer Center Gastric Cancer database","authors":"Lulu Zhao , Penghui Niu , Wanqing Wang , Xue Han , Xiaoyi Luan , Huang Huang , Yawei Zhang , Dongbing Zhao , Jidong Gao , Yingtai Chen","doi":"10.1016/j.jncc.2024.01.007","DOIUrl":"10.1016/j.jncc.2024.01.007","url":null,"abstract":"<div><h3>Objective</h3><p>Accurate prognosis prediction is critical for individualized-therapy making of gastric cancer patients. We aimed to develop and test 6-month, 1-, 2-, 3-, 5-, and 10-year overall survival (OS) and cancer-specific survival (CSS) prediction models for gastric cancer patients following gastrectomy.</p></div><div><h3>Methods</h3><p>We derived and tested Survival Quilts, a machine learning-based model, to develop 6-month, 1-, 2-, 3-, 5-, and 10-year OS and CSS prediction models. Gastrectomy patients in the development set (<em>n</em> = 20,583) and the internal validation set (<em>n</em> = 5,106) were recruited from the Surveillance, Epidemiology, and End Results (SEER) database, while those in the external validation set (<em>n</em> = 6,352) were recruited from the China National Cancer Center Gastric Cancer (NCCGC) database. Furthermore, we selected gastrectomy patients without neoadjuvant therapy as a subgroup to train and test the prognostic models in order to keep the accuracy of tumor-node-metastasis (TNM) stage. Prognostic performances of these OS and CSS models were assessed using the Concordance Index (C-index) and area under the curve (AUC) values.</p></div><div><h3>Results</h3><p>The machine learning model had a consistently high accuracy in predicting 6-month, 1-, 2-, 3-, 5-, and 10-year OS in the SEER development set (C-index = 0.861, 0.832, 0.789, 0.766, 0.740, and 0.709; AUC = 0.784, 0.828, 0.840, 0.849, 0.869, and 0.902, respectively), SEER validation set (C-index = 0.782, 0.739, 0.712, 0.698, 0.681, and 0.660; AUC = 0.751, 0.772, 0.767, 0.762, 0.766, and 0.787, respectively), and NCCGC set (C-index = 0.691, 0.756, 0.751, 0.737, 0.722, and 0.701; AUC = 0.769, 0.788, 0.790, 0.790, 0.787, and 0.788, respectively). The model was able to predict 6-month, 1-, 2-, 3-, 5-, and 10-year CSS in the SEER development set (C-index = 0.879, 0.858, 0.820, 0.802, 0.784, and 0.774; AUC = 0.756, 0.827, 0.852, 0.863, 0.874, and 0.884, respectively) and SEER validation set (C-index = 0.790, 0.763, 0.741, 0.729, 0.718, and 0.708; AUC = 0.706, 0.758, 0.767, 0.766, 0.766, and 0.764, respectively). In multivariate analysis, the high-risk group with risk score output by 5-year OS model was proved to be a strong survival predictor both in the SEER development set (hazard ratio [HR] = 14.59, 95% confidence interval [CI]: 1.872–2.774, <em>P</em> < 0.001), SEER validation set (HR = 2.28, 95% CI: 13.089–16.293, <em>P</em> < 0.001), and NCCGC set (HR = 1.98, 95% CI: 1.617–2.437, <em>P</em> <em><</em> 0.001). We further explored the prognostic value of risk score resulted 5-year CSS model of gastrectomy patients, and found that high-risk group remained as an independent CSS factor in the SEER development set (HR = 12.81, 95% CI: 11.568–14.194, <em>P</em> < 0.001) and SEER validation set (HR = 1.61, 95% CI: 1.338–1.935, <em>P</em> < 0.001).</p></div><div><h3>Conclusion</h3><p>Survival Quilts could allow accurate prediction of 6-m","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 2","pages":"Pages 142-152"},"PeriodicalIF":7.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S266700542400019X/pdfft?md5=03ed90ba842276e402ae3a9c6f81bd5f&pid=1-s2.0-S266700542400019X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140270218","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-01DOI: 10.1016/j.jncc.2024.04.005
Nana Guo , Qingxin Zhou , Meng Zhang , Xiaowei Chen , Baoqi Zeng , Shanshan Wu , Hongmei Zeng , Mopei Wang , Fei Ma , Feng Sun
Objective
Circulating tumor DNA (ctDNA) is increasingly being used as a potential prognostic biomarker in cancer patients. We aimed to assess the prognostic value of ctDNA in different subtypes of breast cancer patients throughout the whole treatment cycle.
Materials and methods
PubMed, Web of Science, Embase, Cochrane Library, Scopus, and clinical trials.gov databases were searched from January 2016 to May 2022. The following search terms were used: ctDNA OR circulating tumor DNA AND breast cancer OR breast carcinoma. Only studies written in English were included. The following pre-specified criteria should be met for inclusion: (i) original articles, conference abstracts, etc.; (ii) patients with breast cancer; (iii) ctDNA measurement; and (iv) clinical outcome data such as recurrence-free survival (RFS) and overall survival (OS). The random-effects model was preferred considering the potential heterogeneity across studies. The main outcomes are ctDNA detection rate and postoperative long-term outcomes (RFS and OS).
Results
A total of 24 studies were screened. At every measurement time, the ctDNA detection rate of the HR+ subgroup was similar to that of the HR- subgroup (P = 0.075; P = 0.458; P = 0.744; and P = 0.578), and the ctDNA detection rate of the HER2+ subgroup was similar to that of the HER2- subgroup (P = 0.805; P = 0.271; P = 0.807; and P = 0.703). In the HR+ subgroup, RFS and OS of ctDNA positive patients were similar to those of ctDNA negative patients (P = 0.589 and P = 0.110), while RFS and OS of the ctDNA positive group was significantly shorter than those of the ctDNA negative patients in the HR- subgroup (HR = 4.03, P < 0.001; HR = 3.21, P < 0.001). According to HER grouping, the results were the same as above. In the triple negative breast cancer (TNBC) subgroup, the RFS and OS of ctDNA-positive patients was significantly shorter than of the ctDNA negative patients before and after surgery.
Conclusions
ctDNA was more predictive of recurrence-free survival and overall survival in the HR- subgroup than in the HR+ subgroup, and the same result was showed in the HER2- subgroup vs. HER2+ subgroup. The prognosis of the TNBC subtype is closely related to ctDNA before and after surgery.
目的循环肿瘤 DNA(ctDNA)越来越多地被用作癌症患者潜在的预后生物标志物。我们旨在评估ctDNA在不同亚型乳腺癌患者整个治疗周期中的预后价值。材料和方法检索了2016年1月至2022年5月期间的PubMed、Web of Science、Embase、Cochrane Library、Scopus和clinical trials.gov数据库。使用的检索词如下:ctDNA OR 循环肿瘤 DNA AND 乳腺癌 OR 乳腺癌。仅纳入以英语撰写的研究。纳入研究应符合以下预设标准:(i) 原创文章、会议摘要等;(ii) 乳腺癌患者;(iii) ctDNA 测量;(iv) 无复发生存期(RFS)和总生存期(OS)等临床结果数据。考虑到各研究之间可能存在异质性,因此首选随机效应模型。主要结果为ctDNA检出率和术后长期结果(RFS和OS)。在每个测量时间,HR+亚组的ctDNA检出率与HR-亚组相似(P = 0.075;P = 0.458;P = 0.744;P = 0.578),HER2+亚组的ctDNA检出率与HER2-亚组相似(P = 0.805;P = 0.271;P = 0.807;P = 0.703)。在HR+亚组中,ctDNA阳性患者的RFS和OS与ctDNA阴性患者相似(P = 0.589和P = 0.110),而在HR-亚组中,ctDNA阳性组的RFS和OS明显短于ctDNA阴性患者(HR = 4.03,P <0.001;HR = 3.21,P <0.001)。根据 HER 分组,结果与上述相同。在三阴性乳腺癌(TNBC)亚组中,ctDNA阳性患者手术前后的RFS和OS明显短于ctDNA阴性患者。TNBC亚型的预后与手术前后的ctDNA密切相关。
{"title":"The prognostic role of circulating tumor DNA across breast cancer molecular subtypes: A systematic review and meta-analysis","authors":"Nana Guo , Qingxin Zhou , Meng Zhang , Xiaowei Chen , Baoqi Zeng , Shanshan Wu , Hongmei Zeng , Mopei Wang , Fei Ma , Feng Sun","doi":"10.1016/j.jncc.2024.04.005","DOIUrl":"10.1016/j.jncc.2024.04.005","url":null,"abstract":"<div><h3>Objective</h3><p>Circulating tumor DNA (ctDNA) is increasingly being used as a potential prognostic biomarker in cancer patients. We aimed to assess the prognostic value of ctDNA in different subtypes of breast cancer patients throughout the whole treatment cycle.</p></div><div><h3>Materials and methods</h3><p>PubMed, Web of Science, Embase, Cochrane Library, Scopus, and clinical trials.gov databases were searched from January 2016 to May 2022. The following search terms were used: ctDNA OR circulating tumor DNA AND breast cancer OR breast carcinoma. Only studies written in English were included. The following pre-specified criteria should be met for inclusion: (i) original articles, conference abstracts, etc.; (ii) patients with breast cancer; (iii) ctDNA measurement; and (iv) clinical outcome data such as recurrence-free survival (RFS) and overall survival (OS). The random-effects model was preferred considering the potential heterogeneity across studies. The main outcomes are ctDNA detection rate and postoperative long-term outcomes (RFS and OS).</p></div><div><h3>Results</h3><p>A total of 24 studies were screened. At every measurement time, the ctDNA detection rate of the HR+ subgroup was similar to that of the HR- subgroup (<em>P</em> = 0.075; <em>P</em> = 0.458; <em>P</em> = 0.744; and <em>P</em> = 0.578), and the ctDNA detection rate of the HER2+ subgroup was similar to that of the HER2- subgroup (<em>P</em> = 0.805; <em>P</em> = 0.271; <em>P</em> = 0.807; and <em>P</em> = 0.703). In the HR+ subgroup, RFS and OS of ctDNA positive patients were similar to those of ctDNA negative patients (<em>P</em> = 0.589 and <em>P</em> = 0.110), while RFS and OS of the ctDNA positive group was significantly shorter than those of the ctDNA negative patients in the HR- subgroup (HR = 4.03, <em>P</em> < 0.001; HR = 3.21, <em>P</em> < 0.001). According to HER grouping, the results were the same as above. In the triple negative breast cancer (TNBC) subgroup, the RFS and OS of ctDNA-positive patients was significantly shorter than of the ctDNA negative patients before and after surgery.</p></div><div><h3>Conclusions</h3><p>ctDNA was more predictive of recurrence-free survival and overall survival in the HR- subgroup than in the HR+ subgroup, and the same result was showed in the HER2- subgroup <em>vs</em>. HER2+ subgroup. The prognosis of the TNBC subtype is closely related to ctDNA before and after surgery.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 2","pages":"Pages 153-161"},"PeriodicalIF":7.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000267/pdfft?md5=fd31fa5ddae7c55667f1411404631dd6&pid=1-s2.0-S2667005424000267-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141050898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-01DOI: 10.1016/j.jncc.2024.01.008
Yu Wang , Wenqing Wang , Tao Zhang , Yin Yang , Jianyang Wang , Canjun Li , Xin Xu , Yuqi Wu , Ying Jiang , Jinghao Duan , Luhua Wang , Nan Bi
<div><h3>Background</h3><p>Liquid biopsy-based biomarkers, including circulating tumor DNA (ctDNA) and blood tumor mutational burden (bTMB), are recognized as promising predictors of prognoses and responses to immune checkpoint inhibitors (ICIs), despite insufficient sensitivity of single biomarker detection. This research aims to determine whether the combinatorial utility of longitudinal ctDNA with bTMB analysis could improve the prognostic and predictive effects.</p></div><div><h3>Methods</h3><p>This prospective two-center cohort trial, consisting of discovery and validation datasets, enrolled unresectable locally advanced non-small-cell lung cancer (LA-NSCLC) patients and assigned them to chemoradiotherapy (CRT) or CRT + consolidation ICI cohorts from 2018 to 2022. Blood specimens were collected pretreatment, 4 weeks post-CRT, and at progression to assess bTMB and ctDNA using 486-gene next-generation sequencing. Dynamic ∆bTMB was calculated as post-CRT bTMB minus baseline bTMB levels. Decision curve analyses were performed to calculate Concordance index (C-index).</p></div><div><h3>Results</h3><p>One hundred twenty-eight patients were enrolled. In the discovery dataset (<em>n</em> = 73), patients treated with CRT and consolidation ICI had significantly longer overall survival (OS; median not reached [NR] vs 20.2 months; <em>P</em> < 0.001) and progression-free survival (PFS; median 25.2 vs 11.4 months; <em>P</em> = 0.011) than those without ICI. Longitudinal analysis demonstrated a significant decrease in ctDNA abundance post-CRT (<em>P</em> < 0.001) but a relative increase with disease progression. Post-CRT detectable residual ctDNA correlated with significantly shorter OS (median 18.3 months vs NR; <em>P</em> = 0.001) and PFS (median 7.3 vs 25.2 months; <em>P</em> < 0.001). For patients with residual ctDNA, consolidation ICI brought significantly greater OS (median NR vs 14.8 months; <em>P</em> = 0.005) and PFS (median 13.8 vs 6.2 months; <em>P</em> = 0.028) benefit, but no significant difference for patients with ctDNA clearance. Dynamic ∆bTMB was predictive of prognosis. Patients with residual ctDNA and increased ∆bTMB (∆bTMB > 0) had significantly worse OS (median 9.0 vs 23.0 months vs NR; <em>P</em> < 0.001) and PFS (median 3.4 vs 7.3 vs 25.2 months; <em>P</em> < 0.001). The combinatorial model integrating post-CRT ctDNA with ∆bTMB had optimal predictive effects on OS (C-index = 0.723) and PFS (C-index = 0.693), outperforming individual features. In the independent validation set, we confirmed residual ctDNA predicted poorer PFS (median 50.8 vs 14.3 months; <em>P</em> = 0.026) but identified more consolidation ICI benefit (median NR vs 8.3 months; <em>P</em> = 0.039). The combined model exhibited a stable predictive advantage (C-index = 0.742 for PFS).</p></div><div><h3>Conclusions</h3><p>The multiparameter assay integrating qualitative residual ctDNA testing with quantitative ∆bTMB dynamics improves patient prognostic
背景基于液体活检的生物标记物,包括循环肿瘤DNA(ctDNA)和血液肿瘤突变负荷(bTMB),被认为是预测预后和对免疫检查点抑制剂(ICIs)反应的有前途的指标,尽管单一生物标记物检测的灵敏度不足。这项研究旨在确定纵向ctDNA与bTMB分析的组合效用是否能改善预后和预测效果。方法这项前瞻性双中心队列试验由发现数据集和验证数据集组成,从2018年至2022年招募了不可切除的局部晚期非小细胞肺癌(LA-NSCLC)患者,并将他们分配到化疗放疗(CRT)或CRT+巩固ICI队列中。采集治疗前、CRT 后 4 周和进展期的血液标本,使用 486 基因新一代测序技术评估 bTMB 和 ctDNA。动态ΔbTMB的计算方法为CRT后bTMB减去基线bTMB水平。进行决策曲线分析以计算一致性指数(C-index)。在发现数据集中(n = 73),接受 CRT 和巩固 ICI 治疗的患者的总生存期(OS;中位数未达到 [NR] vs 20.2 个月;P < 0.001)和无进展生存期(PFS;中位数 25.2 vs 11.4 个月;P = 0.011)明显长于未接受 ICI 治疗的患者。纵向分析表明,CRT 后 ctDNA 丰度显著下降(P = 0.001),但随着疾病的进展会相对增加。CRT后可检测到的残留ctDNA与明显缩短的OS(中位18.3个月 vs NR;P = 0.001)和PFS(中位7.3个月 vs 25.2个月;P <;0.001)相关。对于有残留ctDNA的患者,巩固ICI可显著提高OS(中位NR vs 14.8个月;P = 0.005)和PFS(中位13.8 vs 6.2个月;P = 0.028),但对于ctDNA清除的患者则无明显差异。动态ΔbTMB可预测预后。残留ctDNA和∆bTMB增加(∆bTMB > 0)的患者的OS(中位9.0个月 vs 23.0个月 vs NR;P <;0.001)和PFS(中位3.4个月 vs 7.3个月 vs 25.2个月;P <;0.001)明显较差。将 CT 后 ctDNA 与 ∆bTMB 整合在一起的组合模型对 OS(C 指数 = 0.723)和 PFS(C 指数 = 0.693)具有最佳预测效果,优于单个特征。在独立验证集中,我们证实残留 ctDNA 预测的 PFS 较差(中位 50.8 个月 vs 14.3 个月;P = 0.026),但识别出了更巩固的 ICI 益处(中位 NR vs 8.3 个月;P = 0.039)。结论将定性残留ctDNA检测与定量ΔbTMB动态分析相结合的多参数检测方法改善了患者预后风险分层和疗效预测,可用于LA-NSCLC的个性化巩固治疗。
{"title":"Dynamic bTMB combined with residual ctDNA improves survival prediction in locally advanced NSCLC patients with chemoradiotherapy and consolidation immunotherapy","authors":"Yu Wang , Wenqing Wang , Tao Zhang , Yin Yang , Jianyang Wang , Canjun Li , Xin Xu , Yuqi Wu , Ying Jiang , Jinghao Duan , Luhua Wang , Nan Bi","doi":"10.1016/j.jncc.2024.01.008","DOIUrl":"10.1016/j.jncc.2024.01.008","url":null,"abstract":"<div><h3>Background</h3><p>Liquid biopsy-based biomarkers, including circulating tumor DNA (ctDNA) and blood tumor mutational burden (bTMB), are recognized as promising predictors of prognoses and responses to immune checkpoint inhibitors (ICIs), despite insufficient sensitivity of single biomarker detection. This research aims to determine whether the combinatorial utility of longitudinal ctDNA with bTMB analysis could improve the prognostic and predictive effects.</p></div><div><h3>Methods</h3><p>This prospective two-center cohort trial, consisting of discovery and validation datasets, enrolled unresectable locally advanced non-small-cell lung cancer (LA-NSCLC) patients and assigned them to chemoradiotherapy (CRT) or CRT + consolidation ICI cohorts from 2018 to 2022. Blood specimens were collected pretreatment, 4 weeks post-CRT, and at progression to assess bTMB and ctDNA using 486-gene next-generation sequencing. Dynamic ∆bTMB was calculated as post-CRT bTMB minus baseline bTMB levels. Decision curve analyses were performed to calculate Concordance index (C-index).</p></div><div><h3>Results</h3><p>One hundred twenty-eight patients were enrolled. In the discovery dataset (<em>n</em> = 73), patients treated with CRT and consolidation ICI had significantly longer overall survival (OS; median not reached [NR] vs 20.2 months; <em>P</em> < 0.001) and progression-free survival (PFS; median 25.2 vs 11.4 months; <em>P</em> = 0.011) than those without ICI. Longitudinal analysis demonstrated a significant decrease in ctDNA abundance post-CRT (<em>P</em> < 0.001) but a relative increase with disease progression. Post-CRT detectable residual ctDNA correlated with significantly shorter OS (median 18.3 months vs NR; <em>P</em> = 0.001) and PFS (median 7.3 vs 25.2 months; <em>P</em> < 0.001). For patients with residual ctDNA, consolidation ICI brought significantly greater OS (median NR vs 14.8 months; <em>P</em> = 0.005) and PFS (median 13.8 vs 6.2 months; <em>P</em> = 0.028) benefit, but no significant difference for patients with ctDNA clearance. Dynamic ∆bTMB was predictive of prognosis. Patients with residual ctDNA and increased ∆bTMB (∆bTMB > 0) had significantly worse OS (median 9.0 vs 23.0 months vs NR; <em>P</em> < 0.001) and PFS (median 3.4 vs 7.3 vs 25.2 months; <em>P</em> < 0.001). The combinatorial model integrating post-CRT ctDNA with ∆bTMB had optimal predictive effects on OS (C-index = 0.723) and PFS (C-index = 0.693), outperforming individual features. In the independent validation set, we confirmed residual ctDNA predicted poorer PFS (median 50.8 vs 14.3 months; <em>P</em> = 0.026) but identified more consolidation ICI benefit (median NR vs 8.3 months; <em>P</em> = 0.039). The combined model exhibited a stable predictive advantage (C-index = 0.742 for PFS).</p></div><div><h3>Conclusions</h3><p>The multiparameter assay integrating qualitative residual ctDNA testing with quantitative ∆bTMB dynamics improves patient prognostic","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 2","pages":"Pages 177-187"},"PeriodicalIF":7.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000218/pdfft?md5=949feb9359945cd4e35bb2c25ed9dbe8&pid=1-s2.0-S2667005424000218-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140771120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-01DOI: 10.1016/j.jncc.2023.12.001
Breast Cancer Expert Committee of National Quality Control Center for Cancer;, Breast Cancer Expert Committee of China Anti-Cancer Association;, Cancer Drug Clinical Research Committee of China Anti-Cancer Association
Breast cancer is the most common cancer among women worldwide. It has been estimated that about 416 000 new cases and over 117 000 deaths of breast cancer occurred in China in 2020. Among the new cases of breast cancer diagnosed each year, 3–10% have distant metastasis at the time of initial diagnosis. In addition, approximately 30% of patients with early-stage breast cancer may eventually experience recurrence or metastases. The 5-year survival rate of patients with advanced breast cancer is only 20% with a median overall survival of 2–3 years. Although advanced breast cancer remains incurable at present, new therapeutic options and multidisciplinary treatment could be utilized to alleviate symptoms, improve quality of life, and prolong patients’ survival. The choice of treatment regimens for patients with advanced breast cancer is very important, and the optimal treatment strategy beyond the first- and second-line therapy is often lacking. Herein, the China Advanced Breast Cancer Guideline Panel discussed and summarized recent clinical evidence, updated the guidelines for the diagnosis and treatment of advanced breast cancer based on the 2020 edition, and formulated the “Guidelines for diagnosis and treatment of advanced breast cancer in China (2022 edition)” for clinicians' reference.
{"title":"Guidelines for diagnosis and treatment of advanced breast cancer in China (2022 edition)","authors":"Breast Cancer Expert Committee of National Quality Control Center for Cancer;, Breast Cancer Expert Committee of China Anti-Cancer Association;, Cancer Drug Clinical Research Committee of China Anti-Cancer Association","doi":"10.1016/j.jncc.2023.12.001","DOIUrl":"10.1016/j.jncc.2023.12.001","url":null,"abstract":"<div><p>Breast cancer is the most common cancer among women worldwide. It has been estimated that about 416 000 new cases and over 117 000 deaths of breast cancer occurred in China in 2020. Among the new cases of breast cancer diagnosed each year, 3–10% have distant metastasis at the time of initial diagnosis. In addition, approximately 30% of patients with early-stage breast cancer may eventually experience recurrence or metastases. The 5-year survival rate of patients with advanced breast cancer is only 20% with a median overall survival of 2–3 years. Although advanced breast cancer remains incurable at present, new therapeutic options and multidisciplinary treatment could be utilized to alleviate symptoms, improve quality of life, and prolong patients’ survival. The choice of treatment regimens for patients with advanced breast cancer is very important, and the optimal treatment strategy beyond the first- and second-line therapy is often lacking. Herein, the China Advanced Breast Cancer Guideline Panel discussed and summarized recent clinical evidence, updated the guidelines for the diagnosis and treatment of advanced breast cancer based on the 2020 edition, and formulated the “Guidelines for diagnosis and treatment of advanced breast cancer in China (2022 edition)” for clinicians' reference.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 2","pages":"Pages 107-127"},"PeriodicalIF":7.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005423000832/pdfft?md5=93a405cc18585713cb1f485c4d294200&pid=1-s2.0-S2667005423000832-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139014771","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-01DOI: 10.1016/j.jncc.2023.11.003
Liyan Xue , Yuan Li , Lili Jiang , Chao Liu , Na Cheng , Changyuan Guo , Yan Jin , Ping Zhou , Xuemin Xue , Yue Wang , Weiya Wang , Yanhui Liu , Jianming Ying
Background
The prediction of response to immunotherapy mostly depends on the programmed death-ligand 1 (PD-L1) immunohistochemistry (IHC) status, and the 22C3 pharmDx assay has been approved in esophageal squamous cell carcinoma (ESCC). However, the widespread use of the 22C3 pharmDx assay is limited due to its availability. Thus, alternative PD-L1 assays are needed. We aimed to investigate the analytical and clinical diagnostic performances of four PD-L1 assays and to compare their concordances with the 22C3 pharmDx assay.
Methods
The PD-L1 22C3 pharmDx assay was performed on the Dako Autostainer Link 48 platform, three testing assays (PD-L1 E1L3N XP antibody [Ab], PD-L1 BP6099 Ab and PD-L1 CST E1L3N Ab) on the Leica BOND-MAX/III platform, and one testing assay (PD-L1 MXR006 Ab) on the Roche VENTANA Benchmark Ultra platform. A total of 218 ESCC cases from four centers were included in this retrospective study. Professionals from each center stained and read the IHC slides independently and determined the combined positive score (CPS) and the tumor proportion score (TPS).
Results
Regarding analytical performance, the four testing assays demonstrated good correlations with the 22C3 pharmDx assay when evaluated by the TPS or CPS (ρ > 0.8 for all four assays). Regarding diagnostic performance (CPS ≥ 10 was used as the cutoff), the four testing assays showed moderate concordances with the 22C3 pharmDx assay (kappa > 0.7 for all four assays). The overall percent agreements between each testing assay and the 22C3 pharmDx assay was at least 87.2 %.
Conclusion
This study provides insight into the potential interchangeability of the four PD-L1 assays with the 22C3 pharmDx assay.
{"title":"Concordance between four PD-L1 immunohistochemical assays and 22C3 pharmDx assay in esophageal squamous cell carcinoma in a multicenter study","authors":"Liyan Xue , Yuan Li , Lili Jiang , Chao Liu , Na Cheng , Changyuan Guo , Yan Jin , Ping Zhou , Xuemin Xue , Yue Wang , Weiya Wang , Yanhui Liu , Jianming Ying","doi":"10.1016/j.jncc.2023.11.003","DOIUrl":"10.1016/j.jncc.2023.11.003","url":null,"abstract":"<div><h3>Background</h3><p>The prediction of response to immunotherapy mostly depends on the programmed death-ligand 1 (PD-L1) immunohistochemistry (IHC) status, and the 22C3 pharmDx assay has been approved in esophageal squamous cell carcinoma (ESCC). However, the widespread use of the 22C3 pharmDx assay is limited due to its availability. Thus, alternative PD-L1 assays are needed. We aimed to investigate the analytical and clinical diagnostic performances of four PD-L1 assays and to compare their concordances with the 22C3 pharmDx assay.</p></div><div><h3>Methods</h3><p>The PD-L1 22C3 pharmDx assay was performed on the Dako Autostainer Link 48 platform, three testing assays (PD-L1 E1L3N XP antibody [Ab], PD-L1 BP6099 Ab and PD-L1 CST E1L3N Ab) on the Leica BOND-MAX/III platform, and one testing assay (PD-L1 MXR006 Ab) on the Roche VENTANA Benchmark Ultra platform. A total of 218 ESCC cases from four centers were included in this retrospective study. Professionals from each center stained and read the IHC slides independently and determined the combined positive score (CPS) and the tumor proportion score (TPS).</p></div><div><h3>Results</h3><p>Regarding analytical performance, the four testing assays demonstrated good correlations with the 22C3 pharmDx assay when evaluated by the TPS or CPS (ρ > 0.8 for all four assays). Regarding diagnostic performance (CPS ≥ 10 was used as the cutoff), the four testing assays showed moderate concordances with the 22C3 pharmDx assay (kappa > 0.7 for all four assays). The overall percent agreements between each testing assay and the 22C3 pharmDx assay was at least 87.2 %.</p></div><div><h3>Conclusion</h3><p>This study provides insight into the potential interchangeability of the four PD-L1 assays with the 22C3 pharmDx assay.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 2","pages":"Pages 162-168"},"PeriodicalIF":7.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005423000819/pdfft?md5=416d70d5f83e420162a9782dbf74c963&pid=1-s2.0-S2667005423000819-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139299827","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-01DOI: 10.1016/j.jncc.2024.03.001
Lin Li , Wenqin Xie , Li Zhan , Shaodi Wen , Xiao Luo , Shuangbin Xu , Yantong Cai , Wenli Tang , Qianwen Wang , Ming Li , Zijing Xie , Lin Deng , Hongyuan Zhu , Guangchuang Yu
The evolutionary dynamics of cancer, characterized by its profound heterogeneity, demand sophisticated tools for a holistic understanding. This review delves into tumor phylogenetics, an essential approach bridging evolutionary biology with oncology, offering unparalleled insights into cancer's evolutionary trajectory. We provide an overview of the workflow, encompassing study design, data acquisition, and phylogeny reconstruction. Notably, the integration of diverse data sets emerges as a transformative step, enhancing the depth and breadth of evolutionary insights. With this integrated perspective, tumor phylogenetics stands poised to redefine our understanding of cancer evolution and influence therapeutic strategies.
{"title":"Resolving tumor evolution: a phylogenetic approach","authors":"Lin Li , Wenqin Xie , Li Zhan , Shaodi Wen , Xiao Luo , Shuangbin Xu , Yantong Cai , Wenli Tang , Qianwen Wang , Ming Li , Zijing Xie , Lin Deng , Hongyuan Zhu , Guangchuang Yu","doi":"10.1016/j.jncc.2024.03.001","DOIUrl":"10.1016/j.jncc.2024.03.001","url":null,"abstract":"<div><p>The evolutionary dynamics of cancer, characterized by its profound heterogeneity, demand sophisticated tools for a holistic understanding. This review delves into tumor phylogenetics, an essential approach bridging evolutionary biology with oncology, offering unparalleled insights into cancer's evolutionary trajectory. We provide an overview of the workflow, encompassing study design, data acquisition, and phylogeny reconstruction. Notably, the integration of diverse data sets emerges as a transformative step, enhancing the depth and breadth of evolutionary insights. With this integrated perspective, tumor phylogenetics stands poised to redefine our understanding of cancer evolution and influence therapeutic strategies.</p></div>","PeriodicalId":73987,"journal":{"name":"Journal of the National Cancer Center","volume":"4 2","pages":"Pages 97-106"},"PeriodicalIF":7.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2667005424000206/pdfft?md5=30935fad53e6d15809f0011986581edf&pid=1-s2.0-S2667005424000206-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140280676","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}