1
|
Frost MG, Jensen KJ, Jimenez-Solem E, Qvortrup C, Andersen JAL, Petersen TS. Sex disparities in advanced non-small cell lung cancer survival: A Danish nationwide study. Lung Cancer 2025; 202:108485. [PMID: 40058138 DOI: 10.1016/j.lungcan.2025.108485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Historically, cancers often exhibit sexual dimorphism. However, following the introduction of immune checkpoint inhibitors and targeted therapies into routine clinical practice, the association of sex on patient and tumour characteristics, as well as survival remains largely unexplored in advanced NSCLC. PATIENTS AND METHODS In this observational study, we identified adults diagnosed with advanced NSCLC from January 1, 2017, to December 31, 2023, using Danish nationwide health registries and clinical databases. We compared females to males across demographic, social, socioeconomic, patient and tumour characteristics (including PDL-1 expression and driver mutations), treatments, and overall survival (OS) from diagnosis and initiation of the first line of treatment (LOT1). Cox proportional hazards regression was employed, adjusting for key covariates in the overall cohort and stratified by stage, histological subtype, PD-L1 expression level, and treatment type. Propensity score matching was conducted as a sensitivity analysis. RESULTS Among 14,635 included individuals, males comprised 50 % (n = 7,322). Patient and tumour characteristics differed between sexes. The crude and adjusted female-to-male mortality hazard ratio (HR) was 0·82 (95 % confidence interval [CI]: 0·79-0·85) and 0·84 (0·81-0·88) from diagnosis and 0·80 (0·76-0·84) and 0·80 (0·75-0·84) from initiation of LOT1. Median OS for females and males was 8·8 months (95 % CI 8·4-9·2) and 7·0 months (6·7-7·3) from diagnosis and 14·1 months (13·5-15·1) and 10·9 months (10·3-11·5) from initiation of LOT1. Stratified analyses revealed that across all strata, females consistently demonstrated either favourable or comparable outcomes compared to males. CONCLUSIONS This study increases our understanding of sex-based disparities in advanced NSCLC. Adjusted analyses addressing previously suggested explanations for sex disparities in survival still demonstrated a longer survival for females, indicating yet-unidentified sex differences with potential clinical implications. Our results underscore the critical role of acknowledging sex as a key variable in oncology trials, research, and clinical practice.
Collapse
Affiliation(s)
- Matilde Grupe Frost
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark; Copenhagen Phase IV Unit (Phase4CPH), Department of Clinical Pharmacology and Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark; University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| | - Kristoffer Jarlov Jensen
- Copenhagen Phase IV Unit (Phase4CPH), Department of Clinical Pharmacology and Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Espen Jimenez-Solem
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark; Copenhagen Phase IV Unit (Phase4CPH), Department of Clinical Pharmacology and Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark; University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | | | | | - Tonny Studsgaard Petersen
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark; University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
2
|
Wang Y, Wang S, Li N. Accelerating the clinical translation of CD70-targeted chimeric antigen receptor-based cell therapies in oncology: A comprehensive clinical investigation panorama analysis based on the Trialtrove database. Cancer Lett 2025; 614:217510. [PMID: 39922452 DOI: 10.1016/j.canlet.2025.217510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Affiliation(s)
- Yuning Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Paltiel O, Ratnasingam S, Lee HP. Are we ignoring sex differences in haematological malignancies? A call for improved reporting. Br J Haematol 2025. [PMID: 40123129 DOI: 10.1111/bjh.20044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
There are clear sex-based differences in the incidence, risk factors and mortality of most haematologic malignancies (HM). Despite known differences in physiology, haematopoiesis, molecular profiles, drug pharmacokinetics, treatment-related toxicities and treatment experience, males and females receive standardized and identical treatment for most HMs. Previous published work has demonstrated disparities in female representation in cancer clinical trials and highlighted a paucity of information on differential treatment outcomes and toxicities by sex. We analysed references of 182 clinical trials which form the basis of recent treatment guidelines from the National Comprehensive Cancer Network and found a minority (17/9.3%) did not report the sex distribution of trial participants. However, a majority (165/90.6%) did not report sex-disaggregated outcomes. Of those that did, 36.5% showed outcome differences by sex. Academic leadership by women in the assessed trials as well as in guidelines committees was disproportionately lower than their representation in the profession. We call on all clinical trials leaders, consortia and guideline builders to include sex-disaggregated data in their analyses, reporting these in a transparent manner (as per regulations mandating such reporting), and for investigators to assess whether aetiological factors differ by sex. These actions will enhance personalized prevention, therapy and follow-up.
Collapse
Affiliation(s)
- Ora Paltiel
- Department of Hematology and Braun School of Public Health and Community Medicine, Faculty of Medicine, Hadassah-Hebrew University, Jerusalem, Israel
| | - Sumita Ratnasingam
- Department of Haematology, University Hospital Geelong, Geelong, Victoria, Australia
- Faculty of Health, Deakin School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Hui-Peng Lee
- Department of Haematology, Flinders Medical Centre & Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
4
|
Zhu C, Zeltser N, Oh J, Li CH, Boutros PC. Sex Differences in the Cancer Proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643396. [PMID: 40166173 PMCID: PMC11957023 DOI: 10.1101/2025.03.14.643396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Proteins play a central role in cancer biology, as are the most common drug targets and biomarkers. Sex influences the proteome in many diseases, ranging from neurological to cardiovascular. In cancer, sex is associated with incidence, progression and therapeutic response, as well as characteristics of the tumour genome and transcriptome. The extent to which sex differences impact the cancer proteome remains largely unknown. To fill this gap, we quantified sex differences across 1,590 proteomes from eight cancer types, identifying 39 genes with sex-differential proteins abundance in adenocarcinomas of the lung and 15 genes in hepatocellular carcinomas, clear cell cancers of the kidney and adenocarcinomas of the pancreas. A subset of these protein differences could be rationalized by sex-differential copy number aberrations. Pathway analysis showed that sex-differential proteins in lung adenocarcinoma were enriched in MYC and E2F target pathways. These findings highlight the modest impact of sex on the cancer proteome, but the very limited power of existing proteomics cohorts for these analyses.
Collapse
Affiliation(s)
- Chenghao Zhu
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Nicole Zeltser
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Jieun Oh
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Constance H. Li
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Chang TG, Park S, Schäffer AA, Jiang P, Ruppin E. Hallmarks of artificial intelligence contributions to precision oncology. NATURE CANCER 2025; 6:417-431. [PMID: 40055572 PMCID: PMC11957836 DOI: 10.1038/s43018-025-00917-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/21/2025] [Indexed: 03/29/2025]
Abstract
The integration of artificial intelligence (AI) into oncology promises to revolutionize cancer care. In this Review, we discuss ten AI hallmarks in precision oncology, organized into three groups: (1) cancer prevention and diagnosis, encompassing cancer screening, detection and profiling; (2) optimizing current treatments, including patient outcome prediction, treatment planning and monitoring, clinical trial design and matching, and developing response biomarkers; and (3) advancing new treatments by identifying treatment combinations, discovering cancer vulnerabilities and designing drugs. We also survey AI applications in interventional clinical trials and address key challenges to broader clinical adoption of AI: data quality and quantity, model accuracy, clinical relevance and patient benefit, proposing actionable solutions for each.
Collapse
Affiliation(s)
- Tian-Gen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Seongyong Park
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A Schäffer
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Fey RM, Billo A, Clister T, Doan KL, Berry EG, Tibbitts DC, Kulkarni RP. Personalization of Cancer Treatment: Exploring the Role of Chronotherapy in Immune Checkpoint Inhibitor Efficacy. Cancers (Basel) 2025; 17:732. [PMID: 40075580 PMCID: PMC11899640 DOI: 10.3390/cancers17050732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
In the era of precision medicine, mounting evidence suggests that the time of therapy administration, or chronotherapy, has a great impact on treatment outcomes. Chronotherapy involves planning treatment timing by considering circadian rhythms, which are 24 h oscillations in behavior and physiology driven by synchronized molecular clocks throughout the body. The value of chronotherapy in cancer treatment is currently under investigation, notably in the effects of treatment timing on efficacy and side effects. Immune checkpoint inhibitor (ICI) therapy is a promising cancer treatment. However, many patients still experience disease progression or need to stop the therapy early due to side effects. There is accumulating evidence that the time of day at which ICI therapy is administered can have a substantial effect on ICI efficacy. Thus, it is important to investigate the intersections of circadian rhythms, chronotherapy, and ICI efficacy. In this review, we provide a brief overview of circadian rhythms in the context of immunity and cancer. Additionally, we outline current applications of chronotherapy for cancer treatment. We synthesize the 29 studies conducted to date that examine the impact of time-of-day administration on the efficacy of ICI therapy, its associated side effects, and sex differences in both efficacy and side effects. We also discuss potential mechanisms underlying these observed results. Finally, we highlight the challenges in this area and future directions for research, including the potential for a chronotherapeutic personalized medicine approach that tailors the time of ICI administration to individual patients' circadian rhythms.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Avery Billo
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Terri Clister
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Khanh L. Doan
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Elizabeth G. Berry
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Deanne C. Tibbitts
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
7
|
Vona R, Cittadini C, Ortona E, Matarrese P. Sex Disparity in Cancer: Role of Autophagy and Estrogen Receptors. Cells 2025; 14:273. [PMID: 39996745 PMCID: PMC11854201 DOI: 10.3390/cells14040273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy, a cellular process essential for maintaining homeostasis, plays a fundamental role in recycling damaged components and in adapting to stress. The dysregulation of autophagy is implicated in numerous human diseases, including cancer, where it exhibits a dual role as both a suppressor and a promoter, depending on the context and the stage of tumor development. The significant sex differences observed in autophagic processes are determined by biological factors, such as genetic makeup and sex hormones. Estrogens, through their interaction with specific receptors, modulate autophagy and influence tumor progression, therapy resistance, and the immune response to tumors. In females, the escape from X inactivation and estrogen signaling may be responsible for the advantages, in terms of lower incidence and longer survival, observed in oncology. Women often show better responses to traditional chemotherapy, while men respond better to immunotherapy. The action of sex hormones on the immune system could contribute to these differences. However, women experience more severe adverse reactions to anticancer drugs. The estrogen/autophagy crosstalk-involved in multiple aspects of the tumor, i.e., development, progression and the response to therapy-deserves an in-depth study, as it could highlight sex-specific mechanisms useful for designing innovative and gender-tailored treatments from the perspective of precision medicine.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| | | | | | - Paola Matarrese
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| |
Collapse
|
8
|
Zhang JW, Zhang N, Lyu Y, Zhang XF. Influence of Sex in the Development of Liver Diseases. Semin Liver Dis 2025. [PMID: 39809453 DOI: 10.1055/a-2516-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The liver is a sexually dimorphic organ. Sex differences in prevalence, progression, prognosis, and treatment prevail in most liver diseases, and the mechanism of how liver diseases act differently among male versus female patients has not been fully elucidated. Biological sex differences in normal physiology and disease arise principally from sex hormones and/or sex chromosomes. Sex hormones contribute to the development and progression of most liver diseases, with estrogen- and androgen-mediated signaling pathways mechanistically involved. In addition, genetic factors in sex chromosomes have recently been found to contribute to the sex disparity of many liver diseases, which might explain, to some extent, the difference in gene expression pattern, immune response, and xenobiotic metabolism between men and women. Although increasing evidence suggests that sex is one of the most important modulators of disease prevalence and outcomes, at present, basic and clinical studies have long been sex unbalanced, with female subjects underestimated. As such, this review focuses on sex disparities of liver diseases and summarizes the current understanding of sex-specific mechanisms, including sex hormones, sex chromosomes, etc. We anticipate that understanding sex-specific pathogenesis will aid in promoting personalized therapies for liver disease among male versus female patients.
Collapse
Affiliation(s)
- Jie-Wen Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Nan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Yi Lyu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
9
|
MacDonald WJ, Purcell C, Pinho-Schwermann M, Stubbs NM, Srinivasan PR, El-Deiry WS. Heterogeneity in Cancer. Cancers (Basel) 2025; 17:441. [PMID: 39941808 PMCID: PMC11816170 DOI: 10.3390/cancers17030441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer heterogeneity is a major challenge in oncology, complicating diagnosis, prognostication, and treatment. The clinical heterogeneity of cancer, which leads to differential treatment outcomes between patients with histopathologically similar cancers, is attributable to molecular diversity manifesting through genetic, epigenetic, transcriptomic, microenvironmental, and host biology differences. Heterogeneity is observed between patients, individual metastases, and within individual lesions. This review discusses clinical implications of heterogeneity, emphasizing need for personalized approaches to overcome challenges posed by cancer's diverse presentations. Understanding of emerging molecular diagnostic and analytical techniques can provide a view into the multidimensional complexity of cancer heterogeneity. With over 90% of cancer-related deaths associated with metastasis, we additionally explore the role heterogeneity plays in treatment resistance and recurrence of metastatic lesions. Molecular insights from next-generation sequencing, single-cell transcriptomics, liquid biopsy technology, and artificial intelligence will facilitate the development of combination therapy regimens that can potentially induce lasting and even curative treatment outcomes.
Collapse
Affiliation(s)
- William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Nolan M. Stubbs
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Praveen R. Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and Brown University Health, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| |
Collapse
|
10
|
Shen X, Zhang Y, Li J, Zhou Y, Butensky S, Zhang Y, Cai Z, DeWan AT, Khan SA, Yan H, Johnson CH, Zhu F. OncoSexome: the landscape of sex-based differences in oncologic diseases. Nucleic Acids Res 2025; 53:D1443-D1459. [PMID: 39535034 PMCID: PMC11701605 DOI: 10.1093/nar/gkae1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/28/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The NIH policy on sex as biological variable (SABV) emphasized the importance of sex-based differences in precision oncology. Over 50% of clinically actionable oncology genes are sex-biased, indicating differences in drug efficacy. Research has identified sex differences in non-reproductive cancers, highlighting the need for comprehensive sex-based cancer data. We therefore developed OncoSexome, a multidimensional knowledge base describing sex-based differences in cancer (https://idrblab.org/OncoSexome/) across four key topics: antineoplastic drugs and responses (SDR), oncology-related biomarkers (SBM), risk factors (SRF) and microbial landscape (SML). SDR covers sex-based differences in 2051 anticancer drugs; SBM describes 12 551 sex-differential biomarkers; SRF illustrates 350 sex-dependent risk factors; SML demonstrates 1386 microbes with sex-differential abundances associated with cancer development. OncoSexome is unique in illuminating multifaceted influences of biological sex on cancer, providing both external and endogenous contributors to cancer development and describing sex-based differences for the broadest oncological classes. Given the increasing global research interest in sex-based differences, OncoSexome is expected to impact future precision oncology practices significantly.
Collapse
Affiliation(s)
- Xinyi Shen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Jiamin Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | | | - Yechi Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
- School of Public Health, Zhejiang University, Hangzhou 310058, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Sajid A Khan
- Yale School of Medicine, Yale University, New Haven 06510, USA
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven 06510, USA
| | - Hong Yan
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
11
|
Schalk E, Seltmann A, Böll B, Giesen N, Grans-Siebel J, Kriege O, Lanznaster J, Minti A, Naendrup JH, Neitz J, Panse J, Schmidt-Hieber M, Seggewiss-Bernhardt R, Teschner D, Weber P, Wille K, von Lilienfeld-Toal M, Hentrich M. Sex-Disaggregated Analysis of Central Venous Catheter-Related Bloodstream Infections in Patients with Cancer. Oncol Res Treat 2024; 48:37-47. [PMID: 39527930 PMCID: PMC11809521 DOI: 10.1159/000542535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Men are generally more susceptible to bacterial infections than women. Central venous catheters (CVCs), often used to administer systemic treatment in patients with cancer, are an important source of infection. However, little is known about sex-specific differences of CVC-related bloodstream infections (CRBSIs) in patients with cancer. This study aimed to compare CRBSIs in men versus women in a large cohort of patients with cancer. METHODS Data were derived from the SECRECY registry including nonselected patients with centrally inserted non-tunneled internal jugular or subclavian vein CVCs in 10 hematology and oncology sites in Germany. Only CRBSIs classified as definite CRBSI (dCRBSI) or probable CRBSI were included, and the combination of both was summarized as dpCRBSI. CVCs were matched 1:1 for underlying disease, anatomic site of CVC insertion, type of CVC dressing, antimicrobial coated CVC, complicated CVC insertion, and CVC in situ time by propensity score matching (PSM). Endpoints were CRBSI rates and incidences in CVCs inserted in men versus women. RESULTS A total of 5,075 CVCs registered from March 2013 to March 2024 were included in the analysis, of which 3,024 comprise the PSM cohort. A total of 1,512 (50.0%) CVCs were inserted in men. Underlying diseases mainly were hematological malignancies (96.4%). While there was no statistically significant difference between men and women in the dCRBSI rate (5.4% vs. 4.1%; p = 0.12) and the dCRBSI incidence (3.8 vs. 2.9/1,000 CVC days; p = 0.11), the rate of dpCRBSI (9.9% vs. 6.7%; p = 0.002) and the dpCRBSI incidence (7.0 vs. 4.7/1,000 CVC days; p = 0.002) were significantly higher in men versus women. The proportion of coagulase-negative staphylococci as causative agent of both dCRBSI and dpCRBSI was higher in men than in women (58.8% vs. 41.2%; p = 0.07 and 61.5% vs. 38.5%; p = 0.002, respectively). A multivariable regression revealed neutropenia as an independent risk factor for dCRBSI and male sex as risk factor for dCRBSI and dpCRBSI. CONCLUSION In patients with hematological malignancies, men have a higher risk of CRBSI than women. This finding may be attributed to the high number of jugular vein-inserted CVCs, which in men may be associated with higher rates of skin colonization than in women. Special preventive measures such as earlier removal of CVCs in men may be studied in future. INTRODUCTION Men are generally more susceptible to bacterial infections than women. Central venous catheters (CVCs), often used to administer systemic treatment in patients with cancer, are an important source of infection. However, little is known about sex-specific differences of CVC-related bloodstream infections (CRBSIs) in patients with cancer. This study aimed to compare CRBSIs in men versus women in a large cohort of patients with cancer. METHODS Data were derived from the SECRECY registry including nonselected patients with centrally inserted non-tunneled internal jugular or subclavian vein CVCs in 10 hematology and oncology sites in Germany. Only CRBSIs classified as definite CRBSI (dCRBSI) or probable CRBSI were included, and the combination of both was summarized as dpCRBSI. CVCs were matched 1:1 for underlying disease, anatomic site of CVC insertion, type of CVC dressing, antimicrobial coated CVC, complicated CVC insertion, and CVC in situ time by propensity score matching (PSM). Endpoints were CRBSI rates and incidences in CVCs inserted in men versus women. RESULTS A total of 5,075 CVCs registered from March 2013 to March 2024 were included in the analysis, of which 3,024 comprise the PSM cohort. A total of 1,512 (50.0%) CVCs were inserted in men. Underlying diseases mainly were hematological malignancies (96.4%). While there was no statistically significant difference between men and women in the dCRBSI rate (5.4% vs. 4.1%; p = 0.12) and the dCRBSI incidence (3.8 vs. 2.9/1,000 CVC days; p = 0.11), the rate of dpCRBSI (9.9% vs. 6.7%; p = 0.002) and the dpCRBSI incidence (7.0 vs. 4.7/1,000 CVC days; p = 0.002) were significantly higher in men versus women. The proportion of coagulase-negative staphylococci as causative agent of both dCRBSI and dpCRBSI was higher in men than in women (58.8% vs. 41.2%; p = 0.07 and 61.5% vs. 38.5%; p = 0.002, respectively). A multivariable regression revealed neutropenia as an independent risk factor for dCRBSI and male sex as risk factor for dCRBSI and dpCRBSI. CONCLUSION In patients with hematological malignancies, men have a higher risk of CRBSI than women. This finding may be attributed to the high number of jugular vein-inserted CVCs, which in men may be associated with higher rates of skin colonization than in women. Special preventive measures such as earlier removal of CVCs in men may be studied in future.
Collapse
Affiliation(s)
- Enrico Schalk
- Department of Hematology Oncology and Cell Therapy, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Alva Seltmann
- Institute for Diversity Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Boris Böll
- Department of Internal Medicine I, University Hospital of Cologne, Cologne, Germany
| | - Nicola Giesen
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch-Hospital Stuttgart, Stuttgart, Germany
| | - Judit Grans-Siebel
- Department of Internal Medicine I, University Hospital of Cologne, Cologne, Germany
| | - Oliver Kriege
- Department of Hematology and Medical Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Julia Lanznaster
- Department of Internal Medicine II, Passau Hospital, Passau, Germany
| | - Antrea Minti
- Department of Oncology, Hematology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Aachen, Germany
| | - Jan-Hendrik Naendrup
- Department of Internal Medicine I, University Hospital of Cologne, Cologne, Germany
| | - Julia Neitz
- Department of Hematology and Oncology, Red Cross Hospital Munich, Munich, Germany
| | - Jens Panse
- Department of Oncology, Hematology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Aachen, Germany
| | - Martin Schmidt-Hieber
- Clinic of Hematology, Oncology, Pneumology, Nephrology and Diabetology, Medical University Lausitz Carl-Thiem (MUL-CT), Cottbus, Germany
| | | | - Daniel Teschner
- Department of Hematology and Medical Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Philipp Weber
- Department of Internal Medicine, Klinikum Mutterhaus der Borromaerinnen, Trier, Germany
| | - Kai Wille
- University Clinic for Hematology, Oncology, Hemostaseology and Palliative Care, University of Bochum, Minden, Germany
| | | | - Marcus Hentrich
- Department of Hematology and Oncology, Red Cross Hospital Munich, Munich, Germany
| |
Collapse
|
12
|
Kim H, Park J, Ahn S, Lee H. The impact of sex/gender-specific funding and editorial policies on biomedical research outcomes: a cross-national analysis (2000-2021). Sci Rep 2024; 14:26599. [PMID: 39496696 PMCID: PMC11535369 DOI: 10.1038/s41598-024-77018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024] Open
Abstract
Reflecting sex and gender characteristics in biomedical research is critical to improving health outcomes and reducing adverse effects from medical treatments. This study investigates the impact of sex/gender-specific funding policies and journal editorial standards on the integration of sex/gender analysis in biomedical research publications. Using data from the United States, Canada, the United Kingdom, and other countries between 2000 and 2021, we assessed how these policies influenced research output in the fields of medicine and life sciences. Our findings show that countries with progressive funding policies and journals promoting sex/gender-based reporting have significantly improved research quality and publication rates. This highlights the importance of coordinated policy efforts and editorial practices in advancing integrated sex/gender research. We recommend continued global efforts from policymakers, funding bodies, and journals to embed sex/gender perspectives in scientific inquiry, ensuring more effective and equitable biomedical advancements.
Collapse
Affiliation(s)
- Heajin Kim
- Korea Center for Gendered Innovations for Science and Technology Research, Seoul, Korea
| | - Jinseo Park
- Center for Global R&D Data Analysis, Korea Institute of Science and Technology Information, Seoul, Korea
| | - Sejung Ahn
- Center for Global R&D Data Analysis, Korea Institute of Science and Technology Information, Seoul, Korea
| | - Heisook Lee
- Korea Center for Gendered Innovations for Science and Technology Research, Seoul, Korea.
| |
Collapse
|
13
|
Ponzi D, Palanza P. Sex is a biologically coherent concept: A response to. Horm Behav 2024; 166:105636. [PMID: 39277911 DOI: 10.1016/j.yhbeh.2024.105636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Affiliation(s)
- Davide Ponzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Paola Palanza
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
14
|
Sauter ER, Butera G, Agurs-Collins T. Long-Term Randomized Controlled Trials of Diet Intervention Reports and Their Impact on Cancer: A Systematic Review. Cancers (Basel) 2024; 16:3296. [PMID: 39409915 PMCID: PMC11475623 DOI: 10.3390/cancers16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Most randomized controlled trials (RCTs) assessing the impact of diet on cancer have been short term (<1 year), mostly evaluating breast cancer survivors. Given the many-year interval that is generally required for an intervention to have an impact on cancer risk or prognosis, as well as the fact that lifestyle strategies such as diet modification frequently fail due to lack of adherence over the long term, we focused this systematic review only on longer-term (≥1 year) intervention reports. Diet intervention reports focused on reducing cancer risk in overweight and obese individuals target caloric restriction (every day, some days, or most hours of each day). METHODS This study is a systematic review of RCTs lasting at least 1 year, testing dietary interventions with a primary or secondary endpoint of cancer or a biomarker linked to cancer. RESULTS Fifty-one reports met our review criteria. Twenty of fifty-one (39%) reports are RCTs where the primary endpoint was cancer or a cancer-related biomarker, while the other reports evaluated reports where cancer or a cancer-related biomarker was a secondary endpoint. Thirteen of twenty (65%) primary reports evaluated isocaloric, and the remaining eight evaluated low-calorie diets. All but one of the primary and two secondary isocaloric diet reports evaluated the benefit of a low-fat diet (LFD), with the other three evaluating a Mediterranean diet (MedD). More LCD vs. isocaloric diet primary reports (71% vs. 38%) demonstrated cancer or cancer-related biomarker benefit; the difference in chance of benefit with secondary reports was 85% for LCD vs. 73% for isocaloric diets. Three of three MedD reports demonstrated benefit. Sixty-nine percent (20/29) of the secondary reports came from two large reports: the WHI diet modification trial (15 secondary reports) and the polyp prevention trial (5 secondary reports). Nineteen of twenty-two (86%) primary reports enrolled only women, and three enrolled both men and women. No study that met our criteria enrolled only men, comprising 1447 men in total vs. 62,054 women. Fifteen of twenty (75%) primary reports focus on healthy women or women with breast cancer. Adherence findings are discussed when provided. CONCLUSIONS More long-term RCTs evaluating cancer and cancer-related biomarker endpoints are needed, especially for cancers at sites other than the breast.
Collapse
Affiliation(s)
- Edward R. Sauter
- Division of Cancer Prevention, National Cancer Institute/National Institutes of Health (NIH), 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Gisela Butera
- Office of Research Services, NIH Library, Bethesda, MD 20892, USA;
| | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute/National Institutes of Health (NIH), 9609 Medical Center Drive, Rockville, MD 20850, USA;
| |
Collapse
|
15
|
De Lorenzo A, Itani L, El Ghoch M, Gualtieri P, Frank G, Raffaelli G, Pellegrini M, Di Renzo L. Difference in Body Composition Patterns between Age Groups in Italian Individuals with Overweight and Obesity: When BMI Becomes a Misleading Tool in Nutritional Settings. Nutrients 2024; 16:2415. [PMID: 39125296 PMCID: PMC11314337 DOI: 10.3390/nu16152415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Little is known about the changes in body composition (BC) in people with overweight or obesity. The aim of this study was to assess the differences in BC patterns in this population based on gender and age. A total of 2844 Italian adults of mixed gender and a body mass index (BMI) of ≥25 kg/m2 underwent a BC assessment by means of dual-energy X-ray absorptiometry (DXA). The sample was categorized into three age groups: 'young' (20-39 years), 'middle' (40-59 years), and 'older' (60-80 years) adults, after being matched by body weight and BMI. Males showed higher total body fat percentage (BF%) and a lower total lean mass (LM), progressively from the young to the middle to the older age groups, while females showed similar values for these total compartments between the three age groups. However, in both genders, participants in the middle and older groups were more likely to have a higher trunk fat percentage by +1.23% to +4.21%, and lower appendicular lean mass (ALM) by -0.81 kg to -2.63 kg with respect to the young group, indicating expression of major central adiposity and sarcopenia. While our findings underscore the limitations of BMI to detect these differences between age groups, the identification of new tools suitable for this aim is greatly needed in this population. Moreover, further investigation that clarifies the impact of these differences in BC patterns between gender and age groups on health outcomes is also required.
Collapse
Affiliation(s)
- Antonino De Lorenzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.D.L.); (L.D.R.)
| | - Leila Itani
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Beirut Arab University, Riad El Solh, Beirut 11072809, Lebanon;
| | - Marwan El Ghoch
- Center for the Study of Metabolism, Body Composition and Lifestyle, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.D.L.); (L.D.R.)
| | - Giulia Frank
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.F.); (G.R.)
| | - Glauco Raffaelli
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.F.); (G.R.)
| | - Massimo Pellegrini
- Center for the Study of Metabolism, Body Composition and Lifestyle, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.D.L.); (L.D.R.)
| |
Collapse
|
16
|
Zhao G, Wang Y, Wang S, Li N. Reporting outcome comparisons by sex in oncology clinical trials. Nat Commun 2024; 15:3051. [PMID: 38594230 PMCID: PMC11004016 DOI: 10.1038/s41467-024-47321-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Affiliation(s)
- Guo Zhao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yuning Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|