1
|
Zhang S, Miao L, Tian X, Yang B, Luo B. Opportunities and challenges of immuno-oncology: A bibliometric analysis from 2014 to 2023. Hum Vaccin Immunother 2025; 21:2440203. [PMID: 39885669 PMCID: PMC11792843 DOI: 10.1080/21645515.2024.2440203] [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: 09/11/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 02/01/2025] Open
Abstract
The emergence of immuno-oncology (IO) has led to revolutionary changes in the field of cancer treatment. Despite notable advancements in this field, a thorough exploration of its full depth and extent has yet to be performed. This study provides a comprehensive overview of publications pertaining to IO. Publications on IO from 2014 to 2023 were retrieved by searching the Web of Science Core Collection database (WoSCC). VOSviewer software and Citespace software were used for the visualized analysis. A total of 1,874 articles have been published in the IO domain. The number of publications and citations has been increasing annually. This study also examines the primary research directions within the field of IO. In conclusion, this study offers a comprehensive overview of the opportunities and challenges associated with IO, illuminating the current status of research and indicating potential future trajectories in this rapidly progressing field. This study provides a comprehensive survey of the current research status and hot spots within the field of IO. It will assist researchers in comprehending the current research emphasis and development trends in this field and offers guidance for future research directions.
Collapse
Affiliation(s)
- Siqi Zhang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Lina Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Tian
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Bingxu Yang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Baoping Luo
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
2
|
Hu D, Zhao J, Wu M, Zhou Y, Lyu B, Xu C, Huang C, Su Z, Zhang H, Guo J, Tang W, Chen G, Li Q. Microbial interactions induce the mutational signature of mismatch repair deficiency in colorectal cancer and associated with EPPK1 mutations. Cancer Lett 2025; 625:217807. [PMID: 40383409 DOI: 10.1016/j.canlet.2025.217807] [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: 11/28/2024] [Revised: 05/12/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
To better understand the impact of microbial interactions on the clonal evolution of colorectal cancer (CRC), we conducted high-resolution profiling of the gut microbiome of 101 treatment-naïve primary CRC patients using nanopore sequencing. We performed an integrated analysis of microbiome and tumor exome data to identify symbiotic microbes that interactively influence the mutational processes and the subsequent clonality of CRC. Our results suggested that Dialister pneumosintes and Fusobacterium animalis were both associated with somatic EPPK1 mutations and promote SBS6 (mismatch repair deficiency, dMMR) activity. Notably, we showed that the symbiotic architecture of Dialister pneumosintes and Fusobacterium animalis undergoes significant changes with the mutational status of EPPK1. In addition, we identified specific metabolic pathways involving key metabolites that potentially mediate microbial interactions in CRC. These findings provide new insights into the interplay between the gut microbiome and the mutation landscape of colorectal cancer, thereby informing the clonal evolution of CRC and new strategies for precision medicine.
Collapse
Affiliation(s)
- Dandan Hu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Jialin Zhao
- Department of Hematology, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Miaoqing Wu
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ying Zhou
- Department of Hematology, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Beile Lyu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361100, China
| | - Chaoqun Xu
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361104, China
| | - Chao Huang
- Department of Hematology, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zixuan Su
- Department of Hematology, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hui Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Jintao Guo
- Department of Scientific Research Management, Weifang People's Hospital, Shandong Second Medical University, Weifang, 261041, Shandong Province, China
| | - Weiwei Tang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China; The School of Clinical Medicine, Fujian Medical University, China
| | - Gong Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China.
| | - Qiyuan Li
- Department of Hematology, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
3
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
4
|
Feng H, Xia Y, Wang W, Xu C, Wang Q, Song Z, Li Z, Yu J, Zhong W, Wang Z, Zhang Y, Liu J, Zhang S, Cai X, Liu A, Li W, Zhan P, Liu H, Lyu T, Miao L, Min L, Lin G, Huang L, Yuan J, Jiang Z, Pu X, Rao C, Lyu D, Yu Z, Li X, Tang C, Zhou C, Mei Q, Guo H, Chu Q, Meng R, Liu X, Wu J, Zhou J, Zhu Z, Pan W, Pang F, Hu M, Wang K, Wu F, Xu B, Xu L, Wang L, Zhu Y, Li J, Xie Y, Lin X, Cai J, Wang L, Du Y, Yao W, Shi X, Niu X, Yuan D, Yao Y, Kang J, Zhang J, Zhang C, Gao W, Huang J, Zhang Y, Sun P, Wang H, Ye M, Wang D, Wang Z, Hao Y, Wang Z, Wan B, Lyu D, Jiao X, Shi L, Lan G, Yang S, Shang Y, Wang Y, Li B, Jin G, Zheng K, Ma J, Li W, Zhang Z, Li Z, Li Y, Liu Z, Ma X, Yang N, Wu L, Wang Q, Wang G, Hong Z, Wang J, Fang M, Fang Y, Zhu X, et alFeng H, Xia Y, Wang W, Xu C, Wang Q, Song Z, Li Z, Yu J, Zhong W, Wang Z, Zhang Y, Liu J, Zhang S, Cai X, Liu A, Li W, Zhan P, Liu H, Lyu T, Miao L, Min L, Lin G, Huang L, Yuan J, Jiang Z, Pu X, Rao C, Lyu D, Yu Z, Li X, Tang C, Zhou C, Mei Q, Guo H, Chu Q, Meng R, Liu X, Wu J, Zhou J, Zhu Z, Pan W, Pang F, Hu M, Wang K, Wu F, Xu B, Xu L, Wang L, Zhu Y, Li J, Xie Y, Lin X, Cai J, Wang L, Du Y, Yao W, Shi X, Niu X, Yuan D, Yao Y, Kang J, Zhang J, Zhang C, Gao W, Huang J, Zhang Y, Sun P, Wang H, Ye M, Wang D, Wang Z, Hao Y, Wang Z, Wan B, Lyu D, Jiao X, Shi L, Lan G, Yang S, Shang Y, Wang Y, Li B, Jin G, Zheng K, Ma J, Li W, Zhang Z, Li Z, Li Y, Liu Z, Ma X, Yang N, Wu L, Wang Q, Wang G, Hong Z, Wang J, Fang M, Fang Y, Zhu X, Shen Y, Wang K, Ren X, Zhang Y, Ma S, Zhang J, Song Y, Fang W, Lu Y. Expert consensus on the diagnosis and treatment of non-small cell lung cancer with MET alteration. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0503. [PMID: 40200811 PMCID: PMC11976709 DOI: 10.20892/j.issn.2095-3941.2024.0503] [Show More Authors] [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: 11/14/2024] [Accepted: 02/17/2025] [Indexed: 04/10/2025] Open
Abstract
Alterations in the mesenchymal-epithelial transition factor (MET) gene are critical drivers of non-small cell lung cancer (NSCLC). In recent years advances in precision therapies targeting MET alterations have significantly expanded treatment options for NSCLC patients. These alterations include MET exon 14 skipping mutations (MET exon 14 skipping), MET gene amplifications, MET point mutations (primarily kinase domain mutations), and MET protein overexpression. Accurate identification of these alterations and appropriate selection of patient populations and targeted therapies are essential for improving clinical outcomes. The East China Lung Cancer Group, Youth Committee (ECLUNG YOUNG, Yangtze River Delta Lung Cancer Cooperation Group) has synthesized insights from China's innovative drug development landscape and clinical practice to formulate an expert consensus on the diagnosis and treatment of NSCLC patients with MET alterations. This consensus addresses key areas, such as optimal testing timing, testing methods, testing strategies, quality control measures, and treatment approaches. By offering standardized recommendations, this guidance aims to streamline diagnostic and therapeutic processes and enhance clinical decision-making for NSCLC with MET alterations.
Collapse
Affiliation(s)
- Huijing Feng
- Department of Thoracic Oncology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan 030032, China
| | - Yang Xia
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Wenxian Wang
- Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Chunwei Xu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Zhengbo Song
- Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Ziming Li
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jinpu Yu
- Department of Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou 510080, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jingjing Liu
- Department of Thoracic Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Shirong Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiuyu Cai
- Department of VIP Inpatient, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Anwen Liu
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Ping Zhan
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Tangfeng Lyu
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Liyun Miao
- Department of Respiratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Lingfeng Min
- Department of Respiratory Medicine, Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Gen Lin
- Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou 350014, China
| | - Long Huang
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhansheng Jiang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Xingxiang Pu
- Department of Medical Oncology, Lung Cancer and Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Chuangzhou Rao
- Department of Radiotherapy and Chemotherapy, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo 315010, China
| | - Dongqing Lyu
- Department of Pulmonary Medicine, Taizhou Hospital of Wenzhou Medical University, Taizhou 317000, China
| | - Zongyang Yu
- Department of Respiratory Medicine, the 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China
| | - Xiaoyan Li
- Department of Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100700, China
| | - Chuanhao Tang
- Department of Medical Oncology, Peking University International Hospital, Beijing 102206, China
| | - Chengzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease; Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510300, China
| | - Qi Mei
- Department of Thoracic Oncology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan 030032, China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xuewen Liu
- Department of Oncology, the Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jingxun Wu
- Department of Medical Oncology, the First Affiliated Hospital of Medicine, Xiamen University, Xiamen 361003, China
| | - Jin Zhou
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Weiwei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Fei Pang
- Shanghai OrigiMed Co, Ltd, Shanghai 201114, China
| | - Meizhen Hu
- Shanghai OrigiMed Co, Ltd, Shanghai 201114, China
| | - Kai Wang
- Shanghai OrigiMed Co, Ltd, Shanghai 201114, China
| | - Fan Wu
- Menarini Silicon Biosystems SpA, Shanghai 200333, China
| | - Bingwei Xu
- Department of Biotherapy, Cancer Institute, First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ling Xu
- Department of Interventional Pulmonary Diseases, Anhui Chest Hospital, Hefei 230011, China
| | - Liping Wang
- Department of Oncology, Baotou Cancer Hospital, Baotou 014000, China
| | - Youcai Zhu
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, The Third Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Jisheng Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yanru Xie
- Department of Oncology, Lishui Municipal Central Hospital, Lishui 323000, China
| | - Xinqing Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease; Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510300, China
| | - Jing Cai
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Lin Wang
- Department of Pathology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan 030032, China
| | - Yingying Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wang Yao
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510060, China
| | - Xuefei Shi
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, China
| | - Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dongmei Yuan
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yanwen Yao
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Jing Kang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou 510080, China
| | - Jiatao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou 510080, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou 510080, China
| | - Wenbin Gao
- Department of Oncology, The Third Affiliated Hospital of Shenzhen University, Shenzhen 518001, China
| | - Jianhui Huang
- Department of Oncology, Lishui Municipal Central Hospital, Lishui 323000, China
| | - Yinbin Zhang
- Department of Oncology, the Second Affiliated Hospital of Medical College, Xi’an Jiaotong University, Xi’an 710004, China
| | - Pingli Sun
- Department of Pathology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Hong Wang
- Senior Department of Oncology, The 5th Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Dong Wang
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Zhaofeng Wang
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yue Hao
- Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Zheng Wang
- Department of Radiation Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Bing Wan
- Department of Respiratory Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 210002, China
| | - Donglai Lyu
- Department of Clinical Oncology, The 901 Hospital of Joint Logistics Support Force of People Liberation Army, Hefei 230031, China
| | - Xiaodong Jiao
- Department of Medical Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200070, China
| | - Lin Shi
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Gang Lan
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, The Third Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Shengjie Yang
- Department of Thoracic Surgery, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong 675000, China
| | - Yanhong Shang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Yina Wang
- Department of Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Bihui Li
- Department of Oncology, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China
| | - Gang Jin
- Department of Oncology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Kang Zheng
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Jun Ma
- Department of Thoracic Surgery, Shanxi Provincial People’s Hospital/The Fifth Hospital of Shanxi Medical University/The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China
| | - Wenfeng Li
- Department of Radiation Oncology, First Affiliated Hospital of Wenzhou Medical College, Wenzhou 325000, China
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yuan Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhefeng Liu
- Senior Department of Oncology, The 5th Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Lin Wu
- Department of Medical Oncology, Lung Cancer and Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Guansong Wang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Zhuan Hong
- Department of Medical Oncology, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210029, China
| | - Jiandong Wang
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Meiyu Fang
- Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Xixu Zhu
- Department of Radiation Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yi Shen
- Department of Thoracic Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Ke Wang
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 210000, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Yiping Zhang
- Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Shenglin Ma
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Junping Zhang
- Department of Thoracic Oncology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan 030032, China
| | - Yong Song
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yuanzhi Lu
- Department of Clinical Pathology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| |
Collapse
|
5
|
Kamdee K, Roothumnong E, Thongnoppakhun W, Korphaisarn K, Nakthong P, Dungort P, Meesamarnpong C, Wiboontanasarn S, Tansa-Nga W, Punuch K, Pongsuktavorn K, Tititumjariya W, Lertbussarakam C, Wattanarangsan J, Sritun J, Ridchuayrod N, Pithukpakorn M, Suktitipat B. Comprehensive germline and somatic profiling of high-risk Thai breast cancer via next-generation sequencing. Sci Rep 2025; 15:11427. [PMID: 40181060 PMCID: PMC11968900 DOI: 10.1038/s41598-025-95834-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Breast cancer genomic landscapes differ across ethnic groups, yet the somatic profile of Thai breast tumours has remained uncharacterised. This study analysed 1676 high-hereditary-risk Thai breast cancer patients, identified according to National Comprehensive Cancer Network (NCCN) guideline. Germline alterations were assessed in 1370 cases using a custom 36-core cancer panel. Somatic mutations were characterised in formalin-fixed, paraffin-embedded tumour tissues from 180 of the 1676 patients using the 501-gene Oncomine Comprehensive Assay Plus panel. Pathogenic or likely pathogenic (P/LP) variants were detected in 13% of the 1370 germline analyses, with BRCA1 and BRCA2 being the most frequently altered genes. The prevalence of P/LP variants in BRCA1, BRCA2, and PALB2 differed from that observed in other ethnic cohorts. In somatic profiling, TP53 emerged as the most frequently mutated gene, especially in HER2 and TNBC tumours, whereas MAP3K1 and GATA3 were the most frequently mutated genes in the HR+/HER2- tumours. Moreover, hormone-receptor-positive (HR+) tumours showed distinct mutation patterns compared with other ethnicities. Notably, germline carriers exhibited lower PIK3CA mutation rates than non-carriers. These findings advance our understanding of Thai breast cancer genomics and underscore the importance of ethnic diversity in cancer research, offering insights into tailored screening and therapeutic approaches.
Collapse
Affiliation(s)
- Kornyok Kamdee
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Ekkapong Roothumnong
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanna Thongnoppakhun
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Krittiya Korphaisarn
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Panee Nakthong
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Peerawat Dungort
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chutima Meesamarnpong
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supakit Wiboontanasarn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Warisara Tansa-Nga
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kittiporn Punuch
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Khontawan Pongsuktavorn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Warunya Tititumjariya
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Jantanee Wattanarangsan
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jiraporn Sritun
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Numpueng Ridchuayrod
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Bhoom Suktitipat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
6
|
Nakazawa S, Pecci F, Ricciuti B, Gottlieb FH, Facchinetti F, Harada G, Chen MF, Repetto M, Giacomini F, Jiang J, Locquet MA, Makarem M, Alessi JV, Di Federico A, Aldea M, Garbo E, Gandhi MM, Saini A, Haradon D, Bahcall M, Feng WW, Lee JK, Schrock AB, Drilon A, Awad MM, Jänne PA. Activating Mutations in the MET Kinase Domain Co-Occur With Other Driver Oncogenes and Mediate Resistance to Targeted Therapy in NSCLC. J Thorac Oncol 2025:S1556-0864(25)00650-1. [PMID: 40185329 DOI: 10.1016/j.jtho.2025.03.045] [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] [Revised: 03/05/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
INTRODUCTION MET tyrosine kinase domain (TKD) mutations have recently been characterized as de novo oncogenic drivers in NSCLC. Nevertheless, whether activating MET TKD mutations can confer resistance to targeted therapy in non-MET, oncogene-driven NSCLCs remains unclear. METHODS To characterize the genomic features of tumors with MET TKD mutations in oncogene-driven NSCLC, we performed tumor genomic profiling on two different cohorts of patients with NSCLC. Preclinical models of the most frequently observed MET TKD mutations were generated to determine the effect on sensitivity to targeted therapy. Treatment strategies to overcome MET TKD mutation-mediated resistance were further explored. RESULTS Genomic profiling of more than 115,000 patients with NSCLC found that activating MET TKD mutations are prevalent in 0.15% of cases, and that about half of them co-occur with another oncogenic driver, with a differential pattern in co-occurring MET TKD mutations according to the oncogenic alteration. A review of eight cases with sequential genomic testing revealed that the MET TKD mutation was acquired after systemic therapy in 88% of cases, with a potential contribution of APOBEC mutagenesis underlying this process. In vitro, MET TKD mutation conferred resistance to targeted therapy in diverse oncogene-driven models, which could be overcome by combinatorial treatment against both the primary oncogene and the MET TKD mutation. CONCLUSIONS MET TKD mutation can act as an off-target resistance mechanism in diverse oncogene-driven NSCLC. Combination therapy with an effective MET-targeted therapy can potentially overcome MET TKD mutation-mediated resistance.
Collapse
Affiliation(s)
- Seshiru Nakazawa
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Federica Pecci
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Biagio Ricciuti
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Felix H Gottlieb
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Francesco Facchinetti
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Guilherme Harada
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Monica F Chen
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Matteo Repetto
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Flavia Giacomini
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jie Jiang
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marie-Anaïs Locquet
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Maisam Makarem
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joao V Alessi
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alessandro Di Federico
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mihaela Aldea
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Cancer Medicine Department, Gustave Roussy, Villejuif, France; Paris Saclay University, Kremlin-Bicetre, France
| | - Edoardo Garbo
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Malini M Gandhi
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Arushi Saini
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Danielle Haradon
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Magda Bahcall
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William W Feng
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Alexander Drilon
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Mark M Awad
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Pasi A Jänne
- Department of Medical Oncology, Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
7
|
Paragji A, Shastri V, Nasri E, Ligon JA, Elliott LA, Castillo‐Caro P, Lamba JK, Sayour EJ, Seligson ND. Revisiting CDKN2A dysregulation in Ewing sarcoma. Mol Oncol 2025; 19:994-1001. [PMID: 40080912 PMCID: PMC11977643 DOI: 10.1002/1878-0261.70008] [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: 10/21/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Ewing sarcoma (EwS) is a rare and aggressive malignancy, which frequently affects children. One of the few recurrent genomic variants in EwS is genomic copy number deletion of CDKN2A; however, the clinical consequences of dysregulation of CDKN2A in EwS are unclear. In this study, we revisit CDKN2A to investigate its role as a potential prognostic biomarker in EwS using data from EwS pre-clinical models as well as clinical samples from patients with EwS. We demonstrate the potential essentiality of CDKN2A dysregulation and sustained downstream CDK4/CCND1 activity. Finally, we present evidence that high expression of CDKN2A is a negative prognostic biomarker at diagnosis in EwS in three independent datasets. Our data may suggest that the role of CDKN2A may change across the clinical context of EwS, however, further study is necessary to validate the function of CDKN2A expression in EwS.
Collapse
Affiliation(s)
- Anjali Paragji
- Department of Pharmacotherapy and Translational Research, College of PharmacyThe University of FloridaJacksonvilleFLUSA
| | - Vivek Shastri
- Department of Pharmacotherapy and Translational Research, College of PharmacyThe University of FloridaJacksonvilleFLUSA
| | - Elham Nasri
- Department of Pathology, College of MedicineThe University of FloridaGainesvilleFLUSA
| | - John A. Ligon
- Department of Pediatrics, College of MedicineThe University of FloridaGainesvilleFLUSA
| | - Leighton A. Elliott
- Department of Pediatrics, College of MedicineThe University of FloridaGainesvilleFLUSA
| | - Paul Castillo‐Caro
- Department of Pediatrics, College of MedicineThe University of FloridaGainesvilleFLUSA
| | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of PharmacyThe University of FloridaJacksonvilleFLUSA
| | - Elias J. Sayour
- Department of Neurosurgery, College of MedicineThe University of FloridaGainesvilleFLUSA
| | - Nathan D. Seligson
- Department of Pharmacotherapy and Translational Research, College of PharmacyThe University of FloridaJacksonvilleFLUSA
- Department of Pharmacogenomics and Translational ResearchNemours Children's HealthJacksonvilleFLUSA
| |
Collapse
|
8
|
Tavernari D, Borgeaud M, Liu X, Parikh K, Le X, Ciriello G, Addeo A. Decoding the Clinical and Molecular Signatures of EGFR Common, Compound, and Uncommon Mutations in NSCLC: A Brief Report. J Thorac Oncol 2025; 20:500-506. [PMID: 39694414 DOI: 10.1016/j.jtho.2024.12.012] [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: 10/29/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION EGFR mutations are key oncogenic drivers in lung adenocarcinoma (LUAD), predominantly affecting Asian, nonsmoking, and female populations. Although common mutations, such as exon 19 deletions and L858R, respond well to tyrosine kinase inhibitors (TKIs), uncommon EGFR mutations and compound variants exhibit variable treatment responses. This study aims to compare clinical characteristics and molecular profiles of patients with common, uncommon, and compound EGFR mutations, assessing their implications for therapy outcomes. METHODS We analyzed a multi-cohort genomic dataset of 19,163 patients with LUAD (5,212 with EGFR mutations), categorizing mutations into common, uncommon, and compound classes. Patient demographics, mutational signatures, and tumor microenvironment factors were assessed, with particular attention to smoking status and concomitant alterations in KRAS and TP53. Treatment outcomes were analyzed by time under treatment as a surrogate measure of TKI efficacy. RESULTS Uncommon EGFR mutations, comprising 8.9% of EGFR-altered cases, were significantly more frequent among smokers and associated with tobacco-related mutational signatures. Compared with common EGFR-mutant cases, tumors harboring uncommon EGFR mutations reported higher rates of EGFR amplifications, KRAS, and TP53 mutations. Uncommon mutations also exhibited higher tumor mutational burden and distinct transcriptional profiles linked to cell cycle activity. Median time on treatment with TKIs was notably shorter in patients with uncommon mutations (4.1 mo) than those with common and compound mutations (10.9 mo and 12.4 mo, respectively). CONCLUSIONS This study underscores the clinical and molecular heterogeneity of EGFR mutation classes in LUAD, highlighting the unique profile of uncommon mutations, particularly their association with smoking and co-mutations in KRAS and TP53. Comprehensive molecular testing, including next-generation sequencing, is crucial to identify these uncommon mutations and inform therapeutic decisions. Further investigation into the role of immunotherapy in patients with uncommon EGFR mutations is warranted given the tobacco-related molecular signatures and high tumor mutational burden associated with this subgroup.
Collapse
Affiliation(s)
- Daniele Tavernari
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maxime Borgeaud
- Oncology Service, University Hospital Geneva, Geneva, Switzerland
| | - Ximeng Liu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Alfredo Addeo
- Oncology Service, University Hospital Geneva, Geneva, Switzerland.
| |
Collapse
|
9
|
Chang MJ, Stamos DB, Urtis C, Bowers NL, Schmalz LM, Deyo LJ, Porebski MF, Jabir AR, Bunch PM, Lycan TW, Buchanan Doerfler L, Patwa HS, Waltonen JD, Sullivan CA, Browne JD, Zhang W, Porosnicu M. Mutational Profile of Blood and Tumor Tissue and Biomarkers of Response to PD-1 Inhibitors in Patients with Cutaneous Squamous Cell Carcinoma. Cancers (Basel) 2025; 17:1172. [PMID: 40227722 PMCID: PMC11987913 DOI: 10.3390/cancers17071172] [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: 02/03/2025] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cutaneous squamous cell carcinoma (cSCC) harbors one of the most mutated genomes. There are limited data on the genomic profile and its predictive potential for response to immunotherapy with PD-1 inhibitors in cSCC. METHODS This study retrospectively reviewed cSCC patients treated with PD-1 inhibitor monotherapy at a single institution. Clinical characteristics, treatment outcomes, PD-L1 expression, tumor mutation burden (TMB), and genomic profile in tumor and blood were analyzed. Logistic regression and a support vector classifier were used to validate identified biomarkers of significance. RESULTS Twenty-five patients were evaluable for response and had genomics tested in tumor and/or blood. Of the total, 80% of patients achieved an objective response: 40% complete response (CR), 32% partial response (PR) for more than 6 months, and 8% stable disease (SD) for more than 1 year; 20% of patients progressed on treatment. With a median follow-up of 21 months, progression-free survival (PFS) was 28 months in responders vs. 3 months in non-responders (p = 0.00001). Median PD-L1 was 25% in responders vs. 10% in non-responders (p = 0.39). There was no difference in median TMB between responders and non-responders. Eight gene mutations were significantly more frequent in non-responders than in responders: CDK12 (p = 0.005), CTCF (p = 0.033), CTNNB1 (p = 0.033), IGF1R (p = 0.038), IKBKE (p = 0.016), MLH1 (0.033), QKI (p = 0.016), and TIPARP (p = 0.033). A support vector model of these genes classified responders and non-responders with an accuracy of 0.88 in the training data and 1.0 in the testing data. CONCLUSIONS PD-1 inhibitor monotherapy produces an impressive response. Eight gene mutations were significantly more frequent in non-responders. PD-L1 and TMB were inconclusive in predicting treatment response to anti-PD-L1.
Collapse
Affiliation(s)
- Mark J. Chang
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
| | - Daniel B. Stamos
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
| | - Cetin Urtis
- Center for Cancer Genomics and Precision Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.U.); (L.M.S.); (W.Z.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
| | | | - Lauren M. Schmalz
- Center for Cancer Genomics and Precision Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.U.); (L.M.S.); (W.Z.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
| | - Logan J. Deyo
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
| | - Martin F. Porebski
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
| | - Abdur Rahman Jabir
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
| | - Paul M. Bunch
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Thomas W. Lycan
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
| | - Laura Buchanan Doerfler
- Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA;
| | - Hafiz S. Patwa
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
- Department of Otolaryngology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Joshua D. Waltonen
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
- Department of Otolaryngology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Christopher A. Sullivan
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
- Department of Otolaryngology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - J. Dale Browne
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
- Department of Otolaryngology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Wei Zhang
- Center for Cancer Genomics and Precision Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.U.); (L.M.S.); (W.Z.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
| | - Mercedes Porosnicu
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (M.J.C.); (D.B.S.); (L.J.D.); (M.F.P.); (A.R.J.); (T.W.L.J.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA (H.S.P.); (J.D.W.); (C.A.S.); (J.D.B.)
| |
Collapse
|
10
|
Vogler M, Braun Y, Smith VM, Westhoff MA, Pereira RS, Pieper NM, Anders M, Callens M, Vervliet T, Abbas M, Macip S, Schmid R, Bultynck G, Dyer MJ. The BCL2 family: from apoptosis mechanisms to new advances in targeted therapy. Signal Transduct Target Ther 2025; 10:91. [PMID: 40113751 PMCID: PMC11926181 DOI: 10.1038/s41392-025-02176-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The B cell lymphoma 2 (BCL2) protein family critically controls apoptosis by regulating the release of cytochrome c from mitochondria. In this cutting-edge review, we summarize the basic biology regulating the BCL2 family including canonical and non-canonical functions, and highlight milestones from basic research to clinical applications in cancer and other pathophysiological conditions. We review laboratory and clinical development of BH3-mimetics as well as more recent approaches including proteolysis targeting chimeras (PROTACs), antibody-drug conjugates (ADCs) and tools targeting the BH4 domain of BCL2. The first BCL2-selective BH3-mimetic, venetoclax, showed remarkable efficacy with manageable toxicities and has transformed the treatment of several hematologic malignancies. Following its success, several chemically similar BCL2 inhibitors such as sonrotoclax and lisaftoclax are currently under clinical evaluation, alone and in combination. Genetic analysis highlights the importance of BCL-XL and MCL1 across different cancer types and the possible utility of BH3-mimetics targeting these proteins. However, the development of BH3-mimetics targeting BCL-XL or MCL1 has been more challenging, with on-target toxicities including thrombocytopenia for BCL-XL and cardiac toxicities for MCL1 inhibitors precluding clinical development. Tumor-specific BCL-XL or MCL1 inhibition may be achieved by novel targeting approaches using PROTACs or selective drug delivery strategies and would be transformational in many subtypes of malignancy. Taken together, we envision that the targeting of BCL2 proteins, while already a success story of translational research, may in the foreseeable future have broader clinical applicability and improve the treatment of multiple diseases.
Collapse
Affiliation(s)
- Meike Vogler
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Yannick Braun
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Victoria M Smith
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Raquel S Pereira
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Nadja M Pieper
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Marius Anders
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Manon Callens
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Tim Vervliet
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Maha Abbas
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Ralf Schmid
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Institute for Structural and Chemical Biology, University of Leicester, Leicester, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Martin Js Dyer
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
11
|
Zhong X, Ming Z, Xia Q, Wen X, Ye Z, Luo K, Hu H, Zhuling J, Lei J, Wang S, Xiao X, Yan B, Zhang M. One-tube direct detection of double stranded DNA mutations by a mismatch endonuclease I/CRISPR cas12a cascading system. SENSORS AND ACTUATORS B: CHEMICAL 2025; 426:137093. [DOI: 10.1016/j.snb.2024.137093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
|
12
|
Butera A, Amelio I. Deciphering the significance of p53 mutant proteins. Trends Cell Biol 2025; 35:258-268. [PMID: 38960851 DOI: 10.1016/j.tcb.2024.06.003] [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: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
Mutations in the p53 gene compromise its role as guardian of genomic integrity, yielding predominantly missense p53 mutant proteins. The gain-of-function hypothesis has long suggested that these mutant proteins acquire new oncogenic properties; however, recent studies challenge this notion, indicating that targeting these mutants may not impact the fitness of cancer cells. Mounting evidence indicates that tumorigenesis involves a cooperative interplay between driver mutations and cellular state, influenced by developmental stage, external insults, and tissue damage. Consistently, the behavior and properties of p53 mutants are altered by the context. This article aims to provide a balanced summary of the evolving evidence regarding the contribution of p53 mutants in the biology of cancer while contemplating alternative frameworks to decipher the complexity of p53 mutants within their physiological contexts.
Collapse
Affiliation(s)
- Alessio Butera
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
13
|
Shi R, Sun J, Zhou Z, Shi M, Wang X, Gao Z, Zhao T, Li M, Shu Y. Integration of multiple machine learning approaches develops a gene mutation-based classifier for accurate immunotherapy outcomes. NPJ Precis Oncol 2025; 9:54. [PMID: 40011681 DOI: 10.1038/s41698-025-00842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
In addition to traditional biomarkers like PD-(L)1 expression and tumor mutation burden (TMB), more reliable methods for predicting immune checkpoint blockade (ICB) response in cancer patients are urgently needed. This study utilized multiple machine learning approaches on nonsynonymous mutations to identify key mutations that are most significantly correlated to ICB response. We proposed a classifier, Gene mutation-based Predictive Signature (GPS), to categorize patients based on their predicted response and clinical outcomes post-ICB therapy. GPS outperformed conventional predictors when validated in independent cohorts. Multi-omics analysis and multiplex immunohistochemistry (mIHC) revealed insights into tumor immunogenicity, immune responses, and the tumor microenvironment (TME) in lung adenocarcinoma (LUAD) across different GPS groups. Finally, we validated distinct responses of different GPS samples to ICB in an ex-vivo tumor organoid-PBMC co-culture model. Overall, our findings highlight a simple, robust classifier for accurate ICB response prediction, which could reduce costs, shorten testing times, and facilitate clinical implementation.
Collapse
Affiliation(s)
- Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Sun
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meiqi Shi
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zhaojia Gao
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Tianyu Zhao
- Institute and Clinic for Occupational, Social and Environmental Medicine, LMU University Hospital Munich, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, Lueneburg Municipal Hospital, Lueneburg, Germany
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
14
|
Li X, Yang M, Yang L, Dang X, Li X, Li G. Sequencing of high-frequency mutated genes in breast cancer (BRCA) and associated-functions analysis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2025; 18:46-62. [PMID: 40083350 PMCID: PMC11897711 DOI: 10.62347/yode5431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/29/2024] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Mutations or aberrant expression of genes in an organism tend not to be completely random and this cumulative effect predisposes to the development of malignant tumours. This study aims to reveal the possible aberrant expression of high frequency mutated genes, and then to investigate their role in development, prognosis, signalling pathway function and drug resistance in breast cancer. METHODS The mutated genes in breast cancer (BRCA) clinical samples were identified and detected by high-throughput sequencing. High-frequency mutant genes were counted. Gene expression profiles and the relationship with prognosis were analysed throughout TCGA database. qRT-PCR was used to analyse the mRNA levels of the six high-frequency mutant genes in BRCA tissues and cell lines. IHC was used to analyse the protein levels of the six high-frequency mutant genes in BRCA tissues. The linear interaction, single-cell layer clustering status and the influence in immune cell infiltration degree among these six high-frequency mutant genes were analysed by bioinformatics analysis. The STITCH and cMAP datasets were used for high-frequency mutant gene interaction networks, association signalling pathway enrichment and drug-transcriptome analyses. The effects of trastuzumab on the proliferative capacity of breast cancer cells, as well as on the expression of six high-frequency mutated genes were determined by CCK8 assay. RESULTS The genes that were statistically found to have high-frequency mutations in the samples recruited in the present study by high-throughput sequencing analysis included TP53, PIK3CA, NF1, TBX3, BRCA1 and BRCA2. The expression profiles of these genes and the correlation with prognosis were further demonstrated using the TCGA database: the trend in this study was similar to that of BRCA in TCGA. The mRNA and protein expression of these genes showed that the expression of TP53, NF1, TBX3, BRCA1 and BRCA2 was higher in tumor samples than that in normal samples, with an opposite trend for PIK3CA, a similar trend was observed in BRCA cell lines. The protein expressions of TP53, NF1, TBX3, BRCA1 and BRCA2 displayed the same trend by IHC. Other correlation results include 1) the single cell layer clustering of these six genes resulted in significant clustering with few overlapping regions; 2) these six genes showed different degrees of influence on BRCA immune cell infiltration; 3) these six genes showed a significant correlation between each other; 4) the network of each gene had partially overlapping molecules; and 5) the PI3K pathway was a key association pathway in BRCA. Finally, the cell proliferation ability results confirmed the optimal concentration of trastuzumab and its effect on mRNA expression of these six genes.
Collapse
Affiliation(s)
- Xuelian Li
- Department of Medical Oncology, Brunch of Minhang, Fudan University Shanghai Cancer HospitalShanghai, The People’s Republic of China
| | - Mei Yang
- Department of Medical Oncology, Brunch of Minhang, Fudan University Shanghai Cancer HospitalShanghai, The People’s Republic of China
| | - Liyuan Yang
- Department of Medical Oncology, Brunch of Minhang, Fudan University Shanghai Cancer HospitalShanghai, The People’s Republic of China
| | - Xuefei Dang
- Department of Medical Oncology, Brunch of Minhang, Fudan University Shanghai Cancer HospitalShanghai, The People’s Republic of China
| | - Xueqing Li
- Department of Surgery, Fifth People’s Hospital Affiliated to Fudan UniversityShanghai, The People’s Republic of China
| | - Gang Li
- Department of Medical Oncology, Brunch of Minhang, Fudan University Shanghai Cancer HospitalShanghai, The People’s Republic of China
| |
Collapse
|
15
|
Tsilimigras DI, Stecko H, Moris D, Pawlik TM. Genomic Profiling of Biliary Tract Cancers: Comprehensive Assessment of Anatomic and Geographic Heterogeneity, Co‐Alterations and Outcomes. J Surg Oncol 2025. [DOI: 10.1002/jso.28081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 02/05/2025]
Abstract
ABSTRACTBackgroundBiliary tract cancers (BTCs) represent distinct biological and genomic entities. Anatomic and geographic heterogeneity in genomic profiling of BTC subtypes, genomic co‐alterations, and their impact on long‐term outcomes are not well defined.MethodsGenomic data to characterize alterations among patients with BTCs were derived from the AACR GENIE registry (v15.1) and other genomic data sets. Patterns of mutational co‐occurrence, frequency of co‐alterations, and their impact on long‐term outcomes among BTC patients were examined.ResultsAlterations in IDH1 and FGFR2 genes were mostly noted among intrahepatic cholangiocarcinoma (iCCA) samples, TP53, ERBB2/HER2, and SMAD4 mutations were more frequent among gallbladder cancer (GBC) samples while extrahepatic cholangiocarcinoma (eCCA) more commonly harbored KRAS mutations (all Q < 0.001). Alterations in IDH1 and FGFR2 genes were more frequent among iCCA samples from Western vs. Eastern populations, while KRAS, SMAD4, and ERBB2 mutations were more commonly observed among Eastern populations(all Q < 0.05). FGFR2 gene was frequently co‐mutated with BAP1 (log2OR: 1.55, Q < 0.001), while IDH1 gene was commonly co‐mutated with PBRM1 (log2OR: 1.09, Q < 0.001). Co‐alteration rate among patients with IDH1‐mutant iCCAs, FGFR2‐rearranged iCCAs, KRAS‐mutant eCCA, and HER2‐mutant GBCs were 80.8%, 85.2%, 76.7%, and 100%, respectively. Among patients with iCCA and FGFR2 fusions/rearrangements, harboring co‐alterations in the TP53 pathway or PI3K pathway correlated with worse overall survival (OS), while patients with IDH1‐mutant iCCA had worse OS when harboring co‐alterations in the cell cycle pathway.ConclusionsMarked genomic heterogeneity exists among patients with BTCs based on anatomic and geographic location. The overwhelming majority of BTC patients with clinically significant mutations had concurrent genomic co‐alterations. The current study highlights the molecular complexity of BTCs with multiple alterations that commonly co‐exist and could potentially be targeted to treat BTCs.
Collapse
Affiliation(s)
- Diamantis I. Tsilimigras
- Department of Surgery Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center Columbus Ohio USA
| | - Hunter Stecko
- Department of Surgery Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center Columbus Ohio USA
| | - Dimitrios Moris
- Department of Surgery, Duke University Hospital Duke University Durham North Carolina USA
| | - Timothy M. Pawlik
- Department of Surgery Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center Columbus Ohio USA
| |
Collapse
|
16
|
Sridaran D, Mahajan NP. ACK1/TNK2 kinase: molecular mechanisms and emerging cancer therapeutics. Trends Pharmacol Sci 2025; 46:62-77. [PMID: 39721828 DOI: 10.1016/j.tips.2024.11.006] [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: 09/23/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024]
Abstract
Activated CDC42-associated kinase 1 (ACK1), encoded by the TNK2 gene, is a cytoplasmic non-receptor tyrosine kinase whose aberrant activation correlates positively with cancer severity. Recent research has revealed the functional relevance of this oncokinase - it is an epigenetic regulator that drives cancer progression in multiple malignancies. Although ACK1 is an attractive target for therapeutic intervention, incomplete knowledge of its diverse signaling mechanisms and the lack of specific inhibitors have challenged its clinical success. We summarize recent breakthroughs in understanding ACK1 regulation and cellular signaling, and shed light on its immunomodulatory role in balancing T cell activation. We provide a comprehensive overview of preclinical, proof-of-concept studies of potent ACK1-targeting small-molecule inhibitors that are expected to enter clinical trials for cancer patients.
Collapse
Affiliation(s)
- Dhivya Sridaran
- Division of Urologic Surgery, Department of Surgery, Washington University at St. Louis, St. Louis, MO 63110, USA
| | - Nupam P Mahajan
- Division of Urologic Surgery, Department of Surgery, Washington University at St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Cancer Research Building, Washington University at St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
17
|
Xue X, Gajic ZZ, Caragine CM, Legut M, Walker C, Kim JYS, Wang X, Yan RE, Wessels HH, Lu C, Bapodra N, Gürsoy G, Sanjana NE. Paired CRISPR screens to map gene regulation in cis and trans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625752. [PMID: 39651170 PMCID: PMC11623649 DOI: 10.1101/2024.11.27.625752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Recent massively-parallel approaches to decipher gene regulatory circuits have focused on the discovery of either cis -regulatory elements (CREs) or trans -acting factors. Here, we develop a scalable approach that pairs cis - and trans -regulatory CRISPR screens to systematically dissect how the key immune checkpoint PD-L1 is regulated. In human pancreatic ductal adenocarcinoma (PDAC) cells, we tile the PD-L1 locus using ∼25,000 CRISPR perturbations in constitutive and IFNγ-stimulated conditions. We discover 67 enhancer- or repressor-like CREs and show that distal CREs tend to contact the promoter of PD-L1 and related genes. Next, we measure how loss of all ∼2,000 transcription factors (TFs) in the human genome impacts PD-L1 expression and, using this, we link specific TFs to individual CREs and reveal novel PD-L1 regulatory circuits. For one of these regulatory circuits, we confirm the binding of predicted trans -factors (SRF and BPTF) using CUT&RUN and show that loss of either the CRE or TFs potentiates the anti-cancer activity of primary T cells engineered with a chimeric antigen receptor. Finally, we show that expression of these TFs correlates with PD-L1 expression in vivo in primary PDAC tumors and that somatic mutations in TFs can alter response and overall survival in immune checkpoint blockade-treated patients. Taken together, our approach establishes a generalizable toolkit for decoding the regulatory landscape of any gene or locus in the human genome, yielding insights into gene regulation and clinical impact.
Collapse
|
18
|
Dawood S, Sandhir N, Akasheh M, El Khoury M, Otsmane S, Alnassar M, Abulkhair O, Farhat F, Olsen S. Genomic Landscape of Advanced Solid Tumors in Middle East and North Africa Using Circulating Tumor DNA in Routine Clinical Practice. Oncology 2024:1-13. [PMID: 39342926 DOI: 10.1159/000541571] [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: 03/18/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
INTRODUCTION Next-generation sequencing (NGS) of tumor DNA can detect actionable drivers and help guide therapy for patients with advanced-stage cancers. While tissue-based genotyping is considered a standard of care, blood-based genotyping is emerging as a valid alternative. Tumor genomic profiles may vary by region, and data from the Middle East and North Africa (MENA) are not widely available. This study elucidates the genomic landscape of advanced solid cancers in patients from the MENA region by retrospectively analyzing results from NGS circulating tumor DNA (ctDNA) testing. METHODS In routine clinical practice, 926 plasma samples from 767 patients with advanced cancers from the MENA region were profiled using a comprehensive NGS assay (Guardant360®). We conducted a pan-cancer analysis and sub-analyses focusing on lung, breast, and colorectal cancers. RESULTS In the pan-cancer group, TP53 (58.5%), EGFR (20.4%), and KRAS (18.9%) were the most frequently mutated genes. EGFR (10.2%), FGFR1 (4.9%), and PIK3CA (4.9%) showed the most amplifications, while fusions were observed in 2.7% of patients, including ALK, FGFR2, and RET. For lung adenocarcinoma, EGFR (30.5%), KRAS (19.3%), and ERBB2 (4.6%) were the most frequently identified alterations among the genes recommended for evaluation by the National Comprehensive Cancer Network (NCCN). In patients with breast cancer, PIK3CA (35.3%), ESR1 (21.7%), and BRCA1/2 (13.3%) had the most prevalent alterations among NCCN-recommended genes. In colorectal cancer, KRAS (39.0%), NRAS (8.0%), and BRAF (V600E, 4.0%) were the most observed mutations among genes recommended by the NCCN. Comparing this cohort to publicly available Western and Eastern datasets also indicated similarities (including PIK3CA in breast cancer) and variances (including EGFR in lung adenocarcinoma) in key genes of interest in the analyzed cancer types. CONCLUSION Overall, our findings provide insight into the genomic landscape of individuals with advanced solid organ malignancies from the MENA region and support the role of ctDNA in guiding therapeutic decisions.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Department of Medical Oncology, Mediclinic City Hospital, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | | | | | - Maroun El Khoury
- Cancer Care Center, American Hospital Dubai, Dubai, United Arab Emirates
| | - Sonia Otsmane
- Burjeel Medical City Hospital, Abu Dhabi, United Arab Emirates
| | | | | | - Fadi Farhat
- Department of Hematology and Oncology, Hammoud Hospital University Medical Centre, Sidon, Lebanon
| | | |
Collapse
|
19
|
Healy FM, Turner AL, Marensi V, MacEwan DJ. Mediating kinase activity in Ras-mutant cancer: potential for an individualised approach? Front Pharmacol 2024; 15:1441938. [PMID: 39372214 PMCID: PMC11450236 DOI: 10.3389/fphar.2024.1441938] [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: 05/31/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
It is widely acknowledged that there is a considerable number of oncogenic mutations within the Ras superfamily of small GTPases which are the driving force behind a multitude of cancers. Ras proteins mediate a plethora of kinase pathways, including the MAPK, PI3K, and Ral pathways. Since Ras was considered undruggable until recently, pharmacological targeting of pathways downstream of Ras has been attempted to varying success, though drug resistance has often proven an issue. Nuances between kinase pathway activation in the presence of various Ras mutants are thought to contribute to the resistance, however, the reasoning behind activation of different pathways in different Ras mutational contexts is yet to be fully elucidated. Indeed, such disparities often depend on cancer type and disease progression. However, we are in a revolutionary age of Ras mutant targeted therapy, with direct-targeting KRAS-G12C inhibitors revolutionising the field and achieving FDA-approval in recent years. However, these are only beneficial in a subset of patients. Approximately 90% of Ras-mutant cancers are not KRAS-G12C mutant, and therefore raises the question as to whether other distinct amino acid substitutions within Ras may one day be targetable in a similar manner, and indeed whether better understanding of the downstream pathways these various mutants activate could further improve therapy. Here, we discuss the favouring of kinase pathways across an array of Ras-mutant oncogenic contexts and assess recent advances in pharmacological targeting of various Ras mutants. Ultimately, we will examine the utility of individualised pharmacological approaches to Ras-mediated cancer.
Collapse
Affiliation(s)
- Fiona M. Healy
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Amy L. Turner
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Vanessa Marensi
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Chester Medical School, University of Chester, Chester, United Kingdom
| | - David J. MacEwan
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
20
|
Chen G, Yu Y, Qi Y, Li G, Li N, Meng F, Wang W, Shen R. Comparative analysis of PD-L1 expression and molecular alterations in primary versus metastatic lung adenocarcinoma: a real-world study in China. Front Oncol 2024; 14:1393686. [PMID: 39323996 PMCID: PMC11422015 DOI: 10.3389/fonc.2024.1393686] [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: 05/18/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Objectives Programmed death-ligand 1 (PD-L1) is the only Food and Drug Administration-approved biomarker for monitoring response to immune checkpoint inhibitor (ICI) therapy in patients with lung adenocarcinoma. Understanding the nuances of molecular phenotypes, clinical attributes, and PD-L1 expression levels in primary and metastatic lung adenocarcinoma may help predict response to therapy and assist in the clinical management of lung adenocarcinoma. Methods A total of 235 primary and metastatic lesion specimens from patients with non-small cell lung cancer (NSCLC) an institution in Shandong, China were analyzed. PD-L1 expression was assessed by immunohistochemistry using the 22C3 antibody, and the molecular phenotype was determined by next-generation sequencing of 450 genes. The molecular phenotypes of the primary and metastatic lesions were compared. Results Elevated PD-L1 expression was significantly associated with advanced and metastatic disease (P = 0.001). The distribution of PD-L1 expression varied based on the anatomical location, showing a higher frequency of elevated PD-L1 expression in distal metastases than in the primary tumor. Metastatic lesions exhibited a higher proportion of carcinogenic pathway gene alterations and a greater number of DNA damage-repair pathway gene alterations than the primary lesions. Notably, CDKN2A copy number deletions were more prevalent in metastatic lesions than in primary lesions. Clinical data stemming from research conducted at the Memorial Sloan Kettering Cancer Center revealed an association between the absence of CDKN2A expression and a poorer prognosis in stage I lung adenocarcinoma. Conclusion Samples of metastatic tumors exhibited a higher proportion of elevated PD-L1 expression, a greater number of pathway alterations, and a higher occurrence of CDKN2A copy number deletions than primary samples. This highlights the importance of reinforcing the clinical management and follow-up of patients with CDKN2A deficiency, particularly within the subset of stage I lung adenocarcinoma.
Collapse
Affiliation(s)
- Gang Chen
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yang Yu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Youchao Qi
- Department of Thoracic Surgery, The Second People's Hospital of Dezhou City, Dezhou, Shandong, China
| | - Guangxu Li
- Department of Thoracic Surgery, The Second People's Hospital of Dezhou City, Dezhou, Shandong, China
| | - Ning Li
- Department of Radiotherapy, The Second People's Hospital of Dezhou City, Dezhou, Shandong, China
| | - Fande Meng
- Department of Internal Medicine, Changle County Traditional Chinese Medicine (TMC) Hospital, Weifang, Shandong, China
| | - Wujie Wang
- Department of Interventional Medicine, The Second Hospital, Cheeloo College of Medicine, Institute of Tumor Intervention, Shandong University, Jinan, Shandong, China
| | - Rong Shen
- Department of Minimally Invasive Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
21
|
Yoon J, Oh DY. HER2-targeted therapies beyond breast cancer - an update. Nat Rev Clin Oncol 2024; 21:675-700. [PMID: 39039196 DOI: 10.1038/s41571-024-00924-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/24/2024]
Abstract
The receptor tyrosine-kinase HER2 (also known as ErbB2) is a well-established therapeutic target in patients with breast or gastric cancer selected on the basis of HER2 overexpression on immunohistochemistry and/or ERBB2 amplification on in situ hybridization. With advances in cancer molecular profiling and increased implementation of precision medicine approaches into oncology practice, actionable HER2 alterations in solid tumours have expanded to include ERBB2 mutations in addition to traditional HER2 overexpression and ERBB2 amplification. These various HER2 alterations can be found in solid tumour types beyond breast and gastric cancer, although few HER2-targeted therapeutic options have been established for the other tumour types. Nevertheless, during the 5 years since our previous Review on this topic was published in this journal, obvious and fruitful progress in the development of HER2-targeted therapies has been made, including new disease indications, innovative drugs with diverse mechanisms of action and novel frameworks for approval by regulatory authorities. These advances have culminated in the recent histology-agnostic approval of the anti-HER2 antibody-drug conjugate trastuzumab deruxtecan for patients with HER2-overexpressing solid tumours. In this new Review, we provide an update on the current development landscape of HER2-targeted therapies beyond breast cancer, as well as anticipated future HER2-directed treatment strategies to overcome resistance and thereby improve efficacy and patient outcomes.
Collapse
Affiliation(s)
- Jeesun Yoon
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea.
| |
Collapse
|
22
|
He G, Li Y, Zeng Y, Zhang Y, Jiang Q, Zhang Q, Zhu J, Gong J. Advancements in melanoma immunotherapy: the emergence of Extracellular Vesicle Vaccines. Cell Death Discov 2024; 10:374. [PMID: 39174509 PMCID: PMC11341806 DOI: 10.1038/s41420-024-02150-9] [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: 05/27/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024] Open
Abstract
Malignant melanoma represents a particularly aggressive type of skin cancer, originating from the pathological transformation of melanocytes. While conventional interventions such as surgical resection, chemotherapy, and radiation therapy are available, their non-specificity and collateral damage to normal cells has shifted the focus towards immunotherapy as a notable approach. Extracellular vesicles (EVs) are naturally occurring transporters, and are capable of delivering tumor-specific antigens and directly engaging in the immune response. Multiple types of EVs have emerged as promising platforms for melanoma vaccination. The effectiveness of EV-based melanoma vaccines manifests their ability to potentiate the immune response, particularly by activating dendritic cells (DCs) and CD8+ T lymphocytes, through engineering a synergy of antigen presentation and targeted delivery. Here, this review mainly focuses on the construction strategies for EV vaccines from various sources, their effects, and immunological mechanisms in treating melanoma, as well as the shortcomings and future perspectives in this field. These findings will provide novel insights into the innovative exploitation of EV-based vaccines for melanoma immune therapy.
Collapse
Affiliation(s)
- Guijuan He
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yichuan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuyang Zeng
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yong Zhang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiong Jiang
- Department of Pharmacy, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Xianning Medical College, Hubei University of Science & Technology, Xianning, Hubei, China.
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
23
|
Lu B, Liu Y, Yao Y, Zhu D, Zhang X, Dong K, Xu X, Lv D, Zhao Z, Zhang H, Yang X, Fu W, Huang R, Cao J, Chu J, Pan X, Cui X. Unveiling the unique role of TSPAN7 across tumors: a pan-cancer study incorporating retrospective clinical research and bioinformatic analysis. Biol Direct 2024; 19:72. [PMID: 39175035 PMCID: PMC11340126 DOI: 10.1186/s13062-024-00516-8] [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: 04/28/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND TSPAN7 is an important factor in tumor progression. However, the precise function of TSPAN7 and its role in pan-cancer are not clear. METHODS Based on Xinhua cohort incorporating 370 patients with kidney neoplasm, we conducted differential expression analysis by immunohistochemistry between tumor and normal tissues, and explored correlations of TSPAN7 with patients' survival. Subsequently, we conducted a pan-cancer study, and successively employed differential expression analysis, competing endogenous RNA (ceRNA) analysis, protein-protein interaction (PPI) analysis, correlation analysis of TSPAN7 with clinical characteristics, tumor purity, tumor genomics, tumor immunity, and drug sensitivity. Last but not least, gene set enrichment analysis was applied to identify enriched pathways of TSPAN7. RESULTS In Xinhua cohort, TSPAN7 expression was significantly up-regulated (P-value = 0.0019) in tumor tissues of kidney neoplasm patients. High TSPAN7 expression was associated with decreases in overall survival (OS) (P-value = 0.009) and progression-free survival (P-value = 0.009), and it was further revealed as an independent risk factor for OS (P-value = 0.0326, HR = 5.66, 95%CI = 1.155-27.8). In pan-cancer analysis, TSPAN7 expression was down-regulated in most tumors, and it was associated with patients' survival, tumor purity, tumor genomics, tumor immunity, and drug sensitivity. The ceRNA network and PPI network of TSPAN7 were also constructed. Last but not least, the top five enriched pathways of TSPAN7 in various tumors were identified. CONCLUSION TSPAN7 served as a promising biomarker of various tumors, especially kidney neoplasms, and it was closely associated with tumor purity, tumor genomics, tumor immunology, and drug sensitivity in pan-cancer level.
Collapse
Affiliation(s)
- Bingnan Lu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Dawei Zhu
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China
| | - Xiangmin Zhang
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiao Xu
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China
| | - Donghao Lv
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Wenjia Fu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Runzhi Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China.
| | - Jian Chu
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
24
|
Pecci F, Nakazawa S, Ricciuti B, Harada G, Lee JK, Alessi JV, Barrichello A, Vaz VR, Lamberti G, Di Federico A, Gandhi MM, Gazgalis D, Feng WW, Jiang J, Baldacci S, Locquet MA, Gottlieb FH, Chen MF, Lee E, Haradon D, Smokovich A, Voligny E, Nguyen T, Goel VK, Zimmerman Z, Atwal S, Wang X, Bahcall M, Heist RS, Iqbal S, Gandhi N, Elliott A, Vanderwalde AM, Ma PC, Halmos B, Liu SV, Che J, Schrock AB, Drilon A, Jänne PA, Awad MM. Activating Point Mutations in the MET Kinase Domain Represent a Unique Molecular Subset of Lung Cancer and Other Malignancies Targetable with MET Inhibitors. Cancer Discov 2024; 14:1440-1456. [PMID: 38564707 PMCID: PMC11294820 DOI: 10.1158/2159-8290.cd-23-1217] [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: 10/16/2023] [Revised: 02/23/2024] [Accepted: 04/01/2024] [Indexed: 04/04/2024]
Abstract
Activating point mutations in the MET tyrosine kinase domain (TKD) are oncogenic in a subset of papillary renal cell carcinomas. Here, using comprehensive genomic profiling among >600,000 patients, we identify activating MET TKD point mutations as putative oncogenic driver across diverse cancers, with a frequency of ∼0.5%. The most common mutations in the MET TKD defined as oncogenic or likely oncogenic according to OncoKB resulted in amino acid substitutions at positions H1094, L1195, F1200, D1228, Y1230, M1250, and others. Preclinical modeling of these alterations confirmed their oncogenic potential and also demonstrated differential patterns of sensitivity to type I and type II MET inhibitors. Two patients with metastatic lung adenocarcinoma harboring MET TKD mutations (H1094Y, F1200I) and no other known oncogenic drivers achieved confirmed partial responses to a type I MET inhibitor. Activating MET TKD mutations occur in multiple malignancies and may confer clinical sensitivity to currently available MET inhibitors. Significance: The identification of targetable genomic subsets of cancer has revolutionized precision oncology and offers patients treatments with more selective and effective agents. Here, we demonstrate that activating, oncogenic MET tyrosine kinase domain mutations are found across a diversity of cancer types and are responsive to MET tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Federica Pecci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Seshiru Nakazawa
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Guilherme Harada
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | | | - Joao V Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Adriana Barrichello
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Victor R Vaz
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Giuseppe Lamberti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Malini M Gandhi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dimitris Gazgalis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William W Feng
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jie Jiang
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Simon Baldacci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marie-Anaïs Locquet
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Felix H Gottlieb
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Monica F Chen
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Elinton Lee
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Danielle Haradon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anna Smokovich
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Emma Voligny
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tom Nguyen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vikas K Goel
- Turning Point Therapeutics, Bristol Myers Squibb Company, San Diego, California
| | - Zachary Zimmerman
- Turning Point Therapeutics, Bristol Myers Squibb Company, San Diego, California
| | - Sumandeep Atwal
- Turning Point Therapeutics, Bristol Myers Squibb Company, San Diego, California
| | - Xinan Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Magda Bahcall
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Sumaiya Iqbal
- The Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | | - Patrick C Ma
- Penn State Cancer Institute, Penn State College of Medicine, Penn State University, Hershey, Pennsylvania
| | | | | | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Alexander Drilon
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
25
|
Wang X, Hou K, Ricciuti B, Alessi JV, Li X, Pecci F, Dey R, Luo J, Awad MM, Gusev A, Lin X, Johnson BE, Christiani DC. Additional impact of genetic ancestry over race/ethnicity to prevalence of KRAS mutations and allele-specific subtypes in non-small cell lung cancer. HGG ADVANCES 2024; 5:100320. [PMID: 38902927 PMCID: PMC11452329 DOI: 10.1016/j.xhgg.2024.100320] [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: 02/28/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
The KRAS mutation is the most common oncogenic driver in patients with non-small cell lung cancer (NSCLC). However, a detailed understanding of how self-reported race and/or ethnicity (SIRE), genetically inferred ancestry (GIA), and their interaction affect KRAS mutation is largely unknown. Here, we investigated the associations between SIRE, quantitative GIA, and KRAS mutation and its allele-specific subtypes in a multi-ethnic cohort of 3,918 patients from the Boston Lung Cancer Survival cohort and the Chinese OrigiMed cohort with an independent validation cohort of 1,450 patients with NSCLC. This comprehensive analysis included detailed covariates such as age at diagnosis, sex, clinical stage, cancer histology, and smoking status. We report that SIRE is significantly associated with KRAS mutations, modified by sex, with SIRE-Asian patients showing lower rates of KRAS mutation, transversion substitution, and the allele-specific subtype KRASG12C compared to SIRE-White patients after adjusting for potential confounders. Moreover, GIA was found to correlate with KRAS mutations, where patients with a higher proportion of European ancestry had an increased risk of KRAS mutations, especially more transition substitutions and KRASG12D. Notably, among SIRE-White patients, an increase in European ancestry was linked to a higher likelihood of KRAS mutations, whereas an increase in admixed American ancestry was associated with a reduced likelihood, suggesting that quantitative GIA offers additional information beyond SIRE. The association of SIRE, GIA, and their interplay with KRAS driver mutations in NSCLC highlights the importance of incorporating both into population-based cancer research, aiming to refine clinical decision-making processes and mitigate health disparities.
Collapse
Affiliation(s)
- Xinan Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Kangcheng Hou
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, 611 Charles E. Young Drive, Los Angeles, CA, USA
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology and Center for Cancer Genomics, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Joao V Alessi
- Lowe Center for Thoracic Oncology and Center for Cancer Genomics, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC, USA; Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, USA
| | - Federica Pecci
- Lowe Center for Thoracic Oncology and Center for Cancer Genomics, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Rounak Dey
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Jia Luo
- Lowe Center for Thoracic Oncology and Center for Cancer Genomics, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Mark M Awad
- Lowe Center for Thoracic Oncology and Center for Cancer Genomics, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Alexander Gusev
- McGraw/Patterson Center for Population Sciences, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Bruce E Johnson
- Lowe Center for Thoracic Oncology and Center for Cancer Genomics, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, USA.
| |
Collapse
|
26
|
Cao Y, Lan D, Ke X, Zheng W, Zeng J, Niu N, Fu C, Deng W, Jin S. Investigation of RBM10 mutation and its associations with clinical and molecular characteristics in EGFR-mutant and EGFR-wildtype lung adenocarcinoma. Heliyon 2024; 10:e32287. [PMID: 38912481 PMCID: PMC11192990 DOI: 10.1016/j.heliyon.2024.e32287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/21/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024] Open
Abstract
Background RBM10 is commonly mutated in lung adenocarcinoma (LUAD). However, its role in the pathogenesis of LUAD remains undefined. EGFR-mutant LUAD represents a distinct subset of non-small cell lung cancer (NSCLC). The function of RBM10 in tumor pathogenesis is supposed to differ between EGFR-mutant and EGFR-wt LUAD. This study aimed to interrogate the prevalence of RBM10 mutation in a large cohort of Chinese patients with LUAD and investigate the association of RBM10 mutation with clinical and molecular characteristics of EGFR-mutant and EGFR-wt LUAD. Methods Tumor sequencing data from 2848 Chinese patients with LUAD were retrospectively reviewed and analyzed. The prevalence of RBM10 was also compared with other three cohorts: OrigMed (n = 1222), MSKCC (n = 1267), and TCGA (n = 566). The associations of RBM10 mutation with clinical and molecular characteristics were assessed. An external cohort of 182 patients with LUAD who received PD-1 inhibitor were used to investigate the association of RBM10 mutation with clinical outcomes upon immunotherapy. Results Our cohort showed a higher prevalence of RBM10 in EGFR-mutant LUAD than in EGFR-wt LUAD (14.8 % vs. 6.5 %, p < 0.001). The enrichment of RBM10 mutations in EGFR-mutant LUAD was also seen in another Chinese cohort (OrigMed: 14.9 % vs. 7.8 %, p < 0.001), but not in the two western cohorts (MSKCC: 7.4 % vs. 9.5 %, p = 0.272; TCGA: 8.1 % vs. 6.7 %, p = 0.624). RBM10 mutations co-occurred more frequently with EGFR L858R mutations (23.7 %) than with other types of EGFR mutations (19 del: 7.7 %; other: 7.1 % in others, p < 0.001). In EGFR-mutant LUAD, RBM10 mutations were more commonly found in stage I (18.2 %) and II (21.8 %) vs. stage III (9.4 %) and IV (11.3 %) tumors (p < 0.001). The proportion of PD-L1 positive expression in EGFR-mutant LUAD with concomitant RBM10 mutation was not different from that those without RBM10 mutations (41.8 % vs. 47.9 %, p = 0.566). In contrast, RBM10 mutation occurred more frequently in EGFR-wt LUAD at stage II-IV (stage II: 12.0 %, stage III: 8.7 %, stage IV: 6.6 %) than at stage I (2.8 %). EGFR-wt LUAD with concomitant RBM10 mutations had higher proportions of PD-L1 expression positivity (78.9 % vs. 61.9 %, p = 0.014) and higher tumor mutational load (8.97 vs. 2.99 muts/Mb, p < 0.001) than those without. Patients with EGFR-wt LUAD who also harbored RBM10 loss of function (LOF) mutations had a longer median PFS upon immunotherapy than those with RBM10 non-LOF mutations (7.15 m vs. 2.60 m, HR = 4.83 [1.30-17.94], p = 0.010). Conclusion We comprehensively investigated RBM10 mutations in a Chinese cohort with LUAD. Compared to western cohorts, a significant enrichment of RBM10 mutations in EGFR-mutant LUAD compared to EGFR-wildtype LUAD in the Chinese population. RBM10 mutation shows different associations with clinical and molecular characteristics between EGFR-mutant and EGFR-wt LUAD, suggesting a divergent mechanism between these two subsets via which RBM10 deficiency contribute to tumor pathogenesis. The findings contribute to our understanding of the molecular landscape of LUAD and highlight the importance of considering population-specific factors in cancer genomics research.
Collapse
Affiliation(s)
- Yingyue Cao
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, China
- Department of Immunology, School of Basic Medicine and Forensic Medicine, North Sichuan Medical College, China
| | - Dongmei Lan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Xianni Ke
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Wenyu Zheng
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Jialong Zeng
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Niu Niu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Chunmei Fu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Wencui Deng
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shi Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| |
Collapse
|
27
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [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: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
28
|
Pang LL, Zhou HQ, Zhang YX, Zhuang WT, Pang F, Chen LJ, Liao J, Huang YH, Mao TQ, Mai ZH, Zhang L, Fang WF. SWI/SNF family mutations in advanced NSCLC: genetic characteristics and immune checkpoint inhibitors' therapeutic implication. ESMO Open 2024; 9:103472. [PMID: 38833972 PMCID: PMC11179082 DOI: 10.1016/j.esmoop.2024.103472] [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: 08/27/2023] [Revised: 03/21/2024] [Accepted: 04/21/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND SWItch/Sucrose NonFermentable (SWI/SNF) mutations have garnered increasing attention because of their association with unfavorable prognosis. However, the genetic landscape of SWI/SNF family mutations in Chinese non-small-cell lung cancer (NSCLC) is poorly understood. In addition, the optimal treatment strategy has not yet been determined. PATIENTS AND METHODS We collected sequencing data on 2027 lung tumor samples from multiple centers in China to comprehensively analyze the genomic characteristics of the SWI/SNF family within the Chinese NSCLC population. Meanwhile, 519 patients with NSCLC from Sun Yat-sen University Cancer Center were enrolled to investigate the potential implications of immunotherapy on patients with SWI/SNF mutations and to identify beneficial subpopulations. We also validated our findings in multiple publicly available cohorts. RESULTS Approximately 15% of Chinese patients with lung cancer harbored mutations in the SWI/SNF chromatin remodeling complex, which were mutually exclusive to the EGFR mutations. Patients with SWI/SNFmut NSCLC who received first-line chemoimmunotherapy had better survival outcomes than those who received chemotherapy alone (median progression-free survival: 8.70 versus 6.93 months; P = 0.028). This finding was also confirmed by external validation using the POPLAR/OAK cohort. SWI/SNFmut NSCLC is frequently characterized by high tumor mutational burden and concurrent TP53 or STK11/KEAP mutations. Further analysis indicated that TP53 and STK11/KEAP1 mutations could be stratifying factors in facilitating personalized immunotherapy and guiding patient selection. CONCLUSIONS This study provides a step forward in understanding the genetic and immunological characterization of SWI/SNF genetic alterations. Moreover, our study reveals substantial benefits of immunotherapy over chemotherapy for SWI/SNF-mutant patients, especially the SWI/SNFmut and TP53mut subgroups.
Collapse
Affiliation(s)
- L-L Pang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - H-Q Zhou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - Y-X Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - W-T Zhuang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - F Pang
- Shanghai OrigiMed Co., Ltd, Shanghai
| | - L-J Chen
- Shanghai OrigiMed Co., Ltd, Shanghai
| | - J Liao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - Y-H Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - T-Q Mao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - Z-H Mai
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - L Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou.
| | - W-F Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou.
| |
Collapse
|
29
|
Deng C, Li ZX, Xie CJ, Zhang QL, Hu BS, Wang MD, Mei J, Yang C, Zhong Z, Wang KW. Pan-cancer analysis of CDKN2A alterations identifies a subset of gastric cancer with a cold tumor immune microenvironment. Hum Genomics 2024; 18:55. [PMID: 38822443 PMCID: PMC11143690 DOI: 10.1186/s40246-024-00615-7] [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: 06/28/2023] [Accepted: 05/03/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Although CDKN2A alteration has been explored as a favorable factor for tumorigenesis in pan-cancers, the association between CDKN2A point mutation (MUT) and intragenic deletion (DEL) and response to immune checkpoint inhibitors (ICIs) is still disputed. This study aims to determine the associations of CDKN2A MUT and DEL with overall survival (OS) and response to immune checkpoint inhibitors treatment (ICIs) among pan-cancers and the clinical features of CDKN2A-altered gastric cancer. METHODS This study included 45,000 tumor patients that underwent tumor sequencing across 33 cancer types from four cohorts, the MSK-MetTropism, MSK-IMPACT, OrigiMed2020 and TCGA cohorts. Clinical outcomes and genomic factors associated with response to ICIs, including tumor mutational burden, copy number alteration, neoantigen load, microsatellite instability, tumor immune microenvironment and immune-related gene signatures, were collected in pan-cancer. Clinicopathologic features and outcomes were assessed in gastric cancer. Patients were grouped based on the presence of CDKN2A wild type (WT), CDKN2A MUT, CDKN2A DEL and CDKN2A other alteration (ALT). RESULTS Our research showed that CDKN2A-MUT patients had shorter survival times than CDKN2A-WT patients in the MSK MetTropism and TCGA cohorts, but longer OS in the MSK-IMPACT cohort with ICIs treatment, particularly in patients having metastatic disease. Similar results were observed among pan-cancer patients with CDKN2A DEL and other ALT. Notably, CDKN2A ALT frequency was positively related to tumor-specific objective response rates to ICIs in MSK MetTropism and OrigiMed 2020. Additionally, individuals with esophageal carcinoma or stomach adenocarcinoma who had CDKN2A MUT had poorer OS than patients from the MSK-IMPACT group, but not those with adenocarcinoma. We also found reduced levels of activated NK cells, T cells CD8 and M2 macrophages in tumor tissue from CDKN2A-MUT or DEL pan-cancer patients compared to CDKN2A-WT patients in TCGA cohort. Gastric cancer scRNA-seq data also showed that CDKN2A-ALT cancer contained less CD8 T cells but more exhausted T cells than CDKN2A-WT cancer. A crucial finding of the pathway analysis was the inhibition of three immune-related pathways in the CDKN2A ALT gastric cancer patients, including the interferon alpha response, inflammatory response, and interferon gamma response. CONCLUSIONS This study illustrates the CDKN2A MUT and DEL were associated with a poor outcome across cancers. CDKN2A ALT, on the other hand, have the potential to be used as a biomarker for choosing patients for ICI treatment, notably in esophageal carcinoma and stomach adenocarcinoma.
Collapse
Affiliation(s)
- Chao Deng
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, Jiangsu Province, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zi-Xi Li
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, Jiangsu Province, 214122, China
| | - Chen-Jun Xie
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, Jiangsu Province, 214122, China
| | - Qing-Lin Zhang
- Departments of Gastroenterology, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Ben-Shun Hu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Mei-Dan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, Jiangsu Province, 214122, China
| | - Jie Mei
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Chen Yang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, Jiangsu Province, 214122, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, Macao SAR 999078, China.
| | - Ke-Wei Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, Jiangsu Province, 214122, China.
| |
Collapse
|
30
|
Yu P, Hu C, Ding G, Shi X, Xu J, Cao Y, Chen X, Wu W, Xu Q, Fang J, Huang X, Yuan S, Chen H, Wang Z, Huang L, Pang F, Du Y, Cheng X. Mutation characteristics and molecular evolution of ovarian metastasis from gastric cancer and potential biomarkers for paclitaxel treatment. Nat Commun 2024; 15:3771. [PMID: 38704377 PMCID: PMC11069556 DOI: 10.1038/s41467-024-48144-0] [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: 02/27/2023] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
Ovarian metastasis is one of the major causes of treatment failure in patients with gastric cancer (GC). However, the genomic characteristics of ovarian metastasis in GC remain poorly understood. In this study, we enroll 74 GC patients with ovarian metastasis, with 64 having matched primary and metastatic samples. Here, we show a characterization of the mutation landscape of this disease, alongside an investigation into the molecular heterogeneity and pathway mutation enrichments between synchronous and metachronous metastasis. We classify patients into distinct clonal evolution patterns based on the distribution of mutations in paired samples. Notably, the parallel evolution group exhibits the most favorable prognosis. Additionally, by analyzing the differential response to chemotherapy, we identify potential biomarkers, including SALL4, CCDC105, and CLDN18, for predicting the efficacy of paclitaxel treatment. Furthermore, we validate that CLDN18 fusion mutations improve tumor response to paclitaxel treatment in GC with ovarian metastasis in vitro and vivo.
Collapse
Affiliation(s)
- Pengfei Yu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Guangyu Ding
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | | | - Jingli Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yang Cao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiangliu Chen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wei Wu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Qi Xu
- Department of Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jingquan Fang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xingmao Huang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | | | - Hui Chen
- Shanghai OrigiMed Co., Ltd, Shanghai, PR China
| | | | - Ling Huang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Fei Pang
- Shanghai OrigiMed Co., Ltd, Shanghai, PR China
| | - Yian Du
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
31
|
Horie S, Saito Y, Kogure Y, Mizuno K, Ito Y, Tabata M, Kanai T, Murakami K, Koya J, Kataoka K. Pan-Cancer Comparative and Integrative Analyses of Driver Alterations Using Japanese and International Genomic Databases. Cancer Discov 2024; 14:786-803. [PMID: 38276885 DOI: 10.1158/2159-8290.cd-23-0902] [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: 08/17/2023] [Revised: 12/02/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024]
Abstract
Using 48,627 samples from the Center for Cancer Genomics and Advanced Therapeutics (C-CAT), we present a pan-cancer landscape of driver alterations and their clinical actionability in Japanese patients. Comparison with White patients in Genomics Evidence Neoplasia Information Exchange (GENIE) demonstrates high TP53 mutation frequencies in Asian patients across multiple cancer types. Integration of C-CAT, GENIE, and The Cancer Genome Atlas data reveals many cooccurring and mutually exclusive relationships between driver mutations. At pathway level, mutations in epigenetic regulators frequently cooccur with PI3K pathway molecules. Furthermore, we found significant cooccurring mutations within the epigenetic pathway. Accumulation of mutations in epigenetic regulators causes increased proliferation-related transcriptomic signatures. Loss-of-function of many epigenetic drivers inhibits cell proliferation in their wild-type cell lines, but this effect is attenuated in those harboring mutations of not only the same but also different epigenetic drivers. Our analyses dissect various genetic properties and provide valuable resources for precision medicine in cancer. SIGNIFICANCE We present a genetic landscape of 26 principal cancer types/subtypes, including Asian-prevalent ones, in Japanese patients. Multicohort data integration unveils numerous cooccurring and exclusive relationships between driver mutations, identifying cooccurrence of multiple mutations in epigenetic regulators, which coordinately cause transcriptional and phenotypic changes. These findings provide insights into epigenetic regulator-driven oncogenesis. This article is featured in Selected Articles from This Issue, p. 695.
Collapse
Affiliation(s)
- Sara Horie
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Saito
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yasunori Kogure
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kota Mizuno
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Ito
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Mariko Tabata
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takanori Kanai
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Murakami
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Junji Koya
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Moffat GT, Hu ZI, Meric-Bernstam F, Kong EK, Pavlick D, Ross JS, Murugesan K, Kwong L, De Armas AD, Korkut A, Javle M, Knox JJ. KRAS Allelic Variants in Biliary Tract Cancers. JAMA Netw Open 2024; 7:e249840. [PMID: 38709532 PMCID: PMC11074811 DOI: 10.1001/jamanetworkopen.2024.9840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/06/2024] [Indexed: 05/07/2024] Open
Abstract
Importance Biliary tract cancers (BTCs) contain several actionable molecular alterations, including FGFR2, IDH1, ERBB2 (formerly HER2), and KRAS. KRAS allelic variants are found in 20% to 30% of BTCs, and multiple KRAS inhibitors are currently under clinical investigation. Objectives To describe the genomic landscape, co-sequence variations, immunophenotype, genomic ancestry, and survival outcomes of KRAS-mutated BTCs and to calculate the median overall survival (mOS) for the most common allelic variants. Design, Setting, and Participants This retrospective, multicenter, pooled cohort study obtained clinical and next-generation sequencing data from multiple databases between January 1, 2017, and December 31, 2022. These databases included Princess Margaret Cancer Centre, MD Anderson Cancer Center, Foundation Medicine, American Association for Cancer Research Project GENIE, and cBioPortal for Cancer Genomics. The cohort comprised patients with BTCs who underwent genomic testing. Main Outcome and Measure The main outcome was mOS, defined as date of diagnosis to date of death, which was measured in months. Results A total of 7457 patients (n = 3773 males [50.6%]; mean [SD] age, 63 [5] years) with BTCs and genomic testing were included. Of these patients, 5813 had clinical outcome data available, in whom 1000 KRAS-mutated BTCs were identified. KRAS allelic variants were highly prevalent in perihilar cholangiocarcinoma (28.6%) and extrahepatic cholangiocarcinoma (36.1%). Thirty-six KRAS allelic variants were identified, and the prevalence rates in descending order were G12D (41%), G12V (23%), and Q61H (8%). The variant G12D had the highest mOS of 25.1 (95% CI, 22.0-33.0) months compared with 22.8 (95% CI, 19.6-31.4) months for Q61H and 17.8 (95% CI, 16.3-23.1) months for G12V variants. The majority of KRAS-mutated BTCs (98.9%) were not microsatellite instability-high and had low tumor mutational burden (ranging from a median [IQR] of 1.2 (1.2-2.5) to a mean [SD] of 3.3 [1.3]). Immune profiling through RNA sequencing of KRAS and NRAS-mutated samples showed a pattern toward a more immune-inflamed microenvironment with higher M1 macrophage activation (0.16 vs 0.12; P = .047) and interferon-γ expression compared with wild-type tumors. The G12D variant remained the most common KRAS allelic variant in all patient ancestries. Patients with admixed American ancestry had the highest proportion of G12D variant (45.0%). Conclusions and Relevance This cohort study found that KRAS allelic variants were relatively common and may be potentially actionable genomic alterations in patients with BTCs, especially perihilar cholangiocarcinoma and extrahepatic cholangiocarcinoma. The findings add to the growing data on genomic and immune landscapes of KRAS allelic variants in BTCs and are potentially of value to the planning of specific therapies for this heterogeneous patient group.
Collapse
Affiliation(s)
- Gordon Taylor Moffat
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Zishuo Ian Hu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Funda Meric-Bernstam
- Department of Developmental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston
| | - Elisabeth Kathleen Kong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - Dean Pavlick
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Jeffrey S. Ross
- Foundation Medicine Inc, Cambridge, Massachusetts
- State University of New York Upstate Medical University, Syracuse
| | | | - Lawrence Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Anaemy Danner De Armas
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer J. Knox
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Xu P, Gao Y, Jiang S, Cui Y, Xie Y, Kang Z, Chen YX, Sun D, Fang JY. CHEK2 deficiency increase the response to PD-1 inhibitors by affecting the tumor immune microenvironment. Cancer Lett 2024; 588:216595. [PMID: 38097135 DOI: 10.1016/j.canlet.2023.216595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/13/2023] [Accepted: 12/06/2023] [Indexed: 03/12/2024]
Abstract
Immune checkpoint blockade (ICB) therapy has improved treatment effects in multiple cancers. Gene mutations in the DNA damage repair pathway (DDR) may cause genomic instability and may relate to the efficacy of ICB. Checkpoint kinase 2 (CHEK2) and polymerase epsilon (POLE) are important genes in the DDR. In this study, we aimed to study the impact of CHEK2 deficiency mutations on the response to ICB. We found that tumors with CHEK2 mutations had a significantly higher tumor mutational burden (TMB) compared to those with CHEK2-WT in a pancancer database. We noted that CHEK2 deficiency mutations potentiated the anti-tumor effect of anti-PD-1 therapy in MC38 and B16 tumor-bearing mice with the decrease of tumor volume and tumor weight after anti-PD-1 treatment. Mechanistically, CHEK2 deficiency tumors were with the increased cytotoxic CD8+ T-cell infiltration, especially cytotoxic CD8+ T cells, and modulated the tumor-immune microenvironment with an upregulated immune inflammatory pathway and antigen presentation pathway after anti-PD-1 treatment. Furthermore, murine models with POLE mutations confirmed that CHEK2 deficiency shaped similar mutational and immune landscapes as POLE mutations after anti-PD-1 treatment. Taken together, our results demonstrated that CHEK2 deficiency mutations may increase the response to ICB (eg. anti-PD-1) by influencing the tumor immune microenvironment. This indicated that CHEK2 deficiency mutations were a potentially predictive biomarker and CHEK2 deficiency may potentiate response to immunotherapy.
Collapse
Affiliation(s)
- Pingping Xu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaqi Gao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanshan Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Cui
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanhong Xie
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziran Kang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danfeng Sun
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
34
|
Yao Z, Song P, Jiao W. Pathogenic role of super-enhancers as potential therapeutic targets in lung cancer. Front Pharmacol 2024; 15:1383580. [PMID: 38681203 PMCID: PMC11047458 DOI: 10.3389/fphar.2024.1383580] [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: 02/07/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Lung cancer is still one of the deadliest malignancies today, and most patients with advanced lung cancer pass away from disease progression that is uncontrollable by medications. Super-enhancers (SEs) are large clusters of enhancers in the genome's non-coding sequences that actively trigger transcription. Although SEs have just been identified over the past 10 years, their intricate structure and crucial role in determining cell identity and promoting tumorigenesis and progression are increasingly coming to light. Here, we review the structural composition of SEs, the auto-regulatory circuits, the control mechanisms of downstream genes and pathways, and the characterization of subgroups classified according to SEs in lung cancer. Additionally, we discuss the therapeutic targets, several small-molecule inhibitors, and available treatment options for SEs in lung cancer. Combination therapies have demonstrated considerable advantages in preclinical models, and we anticipate that these drugs will soon enter clinical studies and benefit patients.
Collapse
Affiliation(s)
- Zhiyuan Yao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Song
- Department of Thoracic Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
35
|
Chang J, Zhang Y, Zhou T, Qiao Q, Shan J, Chen Y, Jiang W, Wang Y, Liu S, Wang Y, Yu Y, Li C, Li X. RBM10 C761Y mutation induced oncogenic ASPM isoforms and regulated β-catenin signaling in cholangiocarcinoma. J Exp Clin Cancer Res 2024; 43:104. [PMID: 38576051 PMCID: PMC10993532 DOI: 10.1186/s13046-024-03030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) comprises a heterogeneous group of biliary tract cancer. Our previous CCA mutation pattern study focused on genes in the post-transcription modification process, among which the alternative splicing factor RBM10 captured our attention. However, the roles of RBM10 wild type and mutations in CCA remain unclear. METHODS RBM10 mutation spectrum in CCA was clarified using our initial data and other CCA genomic datasets from domestic and international sources. Real-time PCR and tissue microarray were used to detect RBM10 clinical association. Function assays were conducted to investigate the effects of RBM10 wild type and mutations on CCA. RNA sequencing was to investigate the changes in alternative splicing events in the mutation group compared to the wild-type group. Minigene splicing reporter and interaction assays were performed to elucidate the mechanism of mutation influence on alternative splicing events. RESULTS RBM10 mutations were more common in Chinese CCA populations and exhibited more protein truncation variants. RBM10 exerted a tumor suppressive effect in CCA and correlated with favorable prognosis of CCA patients. The overexpression of wild-type RBM10 enhanced the ASPM exon18 exon skipping event interacting with SRSF2. The C761Y mutation in the C2H2-type zinc finger domain impaired its interaction with SRSF2, resulting in a loss-of-function mutation. Elevated ASPM203 stabilized DVL2 and enhanced β-catenin signaling, which promoted CCA progression. CONCLUSIONS Our results showed that RBM10C761Y-modulated ASPM203 promoted CCA progression in a Wnt/β-catenin signaling-dependent manner. This study may enhance the understanding of the regulatory mechanisms that link mutation-altering splicing variants to CCA.
Collapse
Affiliation(s)
- Jiang Chang
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Yaodong Zhang
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China.
- Key Laboratory for Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
| | - Tao Zhou
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Qian Qiao
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, China
| | - Jijun Shan
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Yananlan Chen
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Wangjie Jiang
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
- Key Laboratory for Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yirui Wang
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Shuochen Liu
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Yuming Wang
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Yue Yu
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
- Key Laboratory for Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Changxian Li
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China.
- Key Laboratory for Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
| | - Xiangcheng Li
- Hepatobiliary Surgery Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China.
- Key Laboratory for Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
36
|
Nguyen Hoang TP, Nguyen TA, Tran NHB, Nguyen Hoang VA, Thi Dao HT, Tran VU, Nguyen YN, Nguyen AT, Nguyen Thi CT, Do Thi TT, Nguyen DS, Nguyen HN, Giang H, Tu LN. Analytical validation and clinical utilization of K-4CARE™: a comprehensive genomic profiling assay with personalized MRD detection. Front Mol Biosci 2024; 11:1334808. [PMID: 38404964 PMCID: PMC10886136 DOI: 10.3389/fmolb.2024.1334808] [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: 11/07/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
Background: Biomarker testing has gradually become standard of care in precision oncology to help physicians select optimal treatment for patients. Compared to single-gene or small gene panel testing, comprehensive genomic profiling (CGP) has emerged as a more time- and tissue-efficient method. This study demonstrated in-depth analytical validation of K-4CARE, a CGP assay that integrates circulating tumor DNA (ctDNA) tracking for residual cancer surveillance. Methods: The assay utilized a panel of 473 cancer-relevant genes with a total length of 1.7 Mb. Reference standards were used to evaluate limit of detection (LOD), concordance, sensitivity, specificity and precision of the assay to detect single nucleotide variants (SNVs), small insertion/deletions (Indels), gene amplification and fusion, microsatellite instability (MSI) and tumor mutational burden (TMB). The assay was then benchmarked against orthogonal methods using 155 clinical samples from 10 cancer types. In selected cancers, top tumor-derived somatic mutations, as ranked by our proprietary algorithm, were used to detect ctDNA in the plasma. Results: For detection of somatic SNVs and Indels, gene fusion and amplification, the assay had sensitivity of >99%, 94% and >99% respectively, and specificity of >99%. Detection of germline variants also achieved sensitivity and specificity of >99%. For TMB measurement, the correlation coefficient between whole-exome sequencing and our targeted panel was 97%. MSI analysis when benchmarked against polymerase chain reaction method showed sensitivity of 94% and specificity of >99%. The concordance between our assay and the TruSight Oncology 500 assay for detection of somatic variants, TMB and MSI measurement was 100%, 89%, and 98% respectively. When CGP-informed mutations were used to personalize ctDNA tracking, the detection rate of ctDNA in liquid biopsy was 79%, and clinical utility in cancer surveillance was demonstrated in 2 case studies. Conclusion: K-4CARE™ assay provides comprehensive and reliable genomic information that fulfills all guideline-based biomarker testing for both targeted therapy and immunotherapy. Integration of ctDNA tracking helps clinicians to further monitor treatment response and ultimately provide well-rounded care to cancer patients.
Collapse
Affiliation(s)
- Thien-Phuc Nguyen Hoang
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Tien Anh Nguyen
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Nam H. B. Tran
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Van-Anh Nguyen Hoang
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Hong Thuy Thi Dao
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Vu-Uyen Tran
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Yen Nhi Nguyen
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Anh Tuan Nguyen
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Cam Tu Nguyen Thi
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | | | - Duy Sinh Nguyen
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Hoai-Nghia Nguyen
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| | - Lan N. Tu
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- Gene Solutions, Ho Chi Minh City, Vietnam
| |
Collapse
|
37
|
Ma C, Teng Q, Shang L, Du F, Li L. Tumor mutation load better predicts the prognosis of patients treated with immune checkpoint inhibitors in upper gastrointestinal cancers: A systematic review and meta-analysis. Cancer Rep (Hoboken) 2024; 7:e1959. [PMID: 38204354 PMCID: PMC10849990 DOI: 10.1002/cnr2.1959] [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: 08/10/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Tumor mutational load (TML) has emerged as a potential biomarker for multiple solid tumors. However, data on its prognostic impact on upper gastrointestinal (UGI) cancer are limited. Therefore, the aim of this systematic review and meta-analysis was to assess the prognostic value of TML for the survival of patients with UGI cancer. METHOD A comprehensive search of the PubMed, Embase, Cochrane Library, and Web of Science databases was conducted up to February 13, 2023. Eleven studies met our inclusion criteria. Hazard ratios (HRs) for progression-free survival and overall survival and their 95% confidence intervals (CIs) were calculated. Subsequently, the combined HR and its 95% CI were calculated for UGI tract cancers in the high and low TML groups. I2 statistics and p-values were used to evaluate heterogeneity. Publication bias, sensitivity, and subgroup analyses were performed to determine sources of heterogeneity. RESULTS In total, 932 patients with UGI tract cancer from 11 publications were included. The high TML group treated with immunotherapy showed significantly improved overall survival (HR = 0.68; 95% CI: 0.53, 0.86; p = .001) and progression-free survival (HR = 0.74; 95% CI: 0.58, 0.95; p = .020) compared with the low TML group. CONCLUSION Our study demonstrated that patients with UGI tumors and higher TML have a better prognosis with immunotherapy, suggesting that TML is a promising predictive biomarker for immunotherapy. REGISTRATION The study protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO Registration No: CRD42023405596).
Collapse
Affiliation(s)
- Chenghao Ma
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Qiong Teng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Fengying Du
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
38
|
Yang X, Lian B, Zhang N, Long J, Li Y, Xue J, Chen X, Wang Y, Wang Y, Xun Z, Piao M, Zhu C, Wang S, Sun H, Song Z, Lu L, Dong X, Wang A, Liu W, Pan J, Hou X, Guan M, Huo L, Shi J, Zhang H, Zhou J, Lu Z, Mao Y, Sang X, Wu L, Yang X, Wang K, Zhao H. Genomic characterization and immunotherapy for microsatellite instability-high in cholangiocarcinoma. BMC Med 2024; 22:42. [PMID: 38281914 PMCID: PMC10823746 DOI: 10.1186/s12916-024-03257-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Microsatellite instability-high (MSI-H) is a unique genomic status in many cancers. However, its role in the genomic features and immunotherapy in cholangiocarcinoma (CCA) is unclear. This study aimed to systematically investigate the genomic characterization and immunotherapy efficacy of MSI-H patients with CCA. METHODS We enrolled 887 patients with CCA in this study. Tumor samples were collected for next-generation sequencing. Differences in genomic alterations between the MSI-H and microsatellite stability (MSS) groups were analyzed. We also investigated the survival of PD-1 inhibitor-based immunotherapy between two groups of 139 patients with advanced CCA. RESULTS Differential genetic alterations between the MSI-H and MSS groups included mutations in ARID1A, ACVR2A, TGFBR2, KMT2D, RNF43, and PBRM1 which were enriched in MSI-H groups. Patients with an MSI-H status have a significantly higher tumor mutation burden (TMB) (median 41.7 vs. 3.1 muts/Mb, P < 0.001) and more positive programmed death ligand 1 (PD-L1) expression (37.5% vs. 11.9%, P < 0.001) than those with an MSS status. Among patients receiving PD-1 inhibitor-based therapy, those with MSI-H had a longer median overall survival (OS, hazard ratio (HR) = 0.17, P = 0.001) and progression-free survival (PFS, HR = 0.14, P < 0.001) than patients with MSS. Integrating MSI-H and PD-L1 expression status (combined positive score ≥ 5) could distinguish the efficacy of immunotherapy. CONCLUSIONS MSI-H status was associated with a higher TMB value and more positive PD-L1 expression in CCA tumors. Moreover, in patients with advanced CCA who received PD-1 inhibitor-based immunotherapy, MSI-H and positive PD-L1 expression were associated with improved both OS and PFS. TRIAL REGISTRATION This study was registered on ClinicalTrials.gov on 07/01/2017 (NCT03892577).
Collapse
Affiliation(s)
- Xu Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Breast Surgery, Peking, Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiran Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingnan Xue
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangqi Chen
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunchao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanyu Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziyu Xun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingjian Piao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenpei Zhu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huishan Sun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | - Jie Pan
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaorong Hou
- Department of Radiotherapy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mei Guan
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jie Shi
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Haohai Zhang
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zhenhui Lu
- Hepatobiliary and Pancreatic Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Yilei Mao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liqun Wu
- Liver Disease Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Kai Wang
- OrigiMed Co., Ltd, Shanghai, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
39
|
Sayin AZ, Abali Z, Senyuz S, Cankara F, Gursoy A, Keskin O. Conformational diversity and protein-protein interfaces in drug repurposing in Ras signaling pathway. Sci Rep 2024; 14:1239. [PMID: 38216592 PMCID: PMC10786864 DOI: 10.1038/s41598-023-50913-8] [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: 08/14/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024] Open
Abstract
We focus on drug repurposing in the Ras signaling pathway, considering structural similarities of protein-protein interfaces. The interfaces formed by physically interacting proteins are found from PDB if available and via PRISM (PRotein Interaction by Structural Matching) otherwise. The structural coverage of these interactions has been increased from 21 to 92% using PRISM. Multiple conformations of each protein are used to include protein dynamics and diversity. Next, we find FDA-approved drugs bound to structurally similar protein-protein interfaces. The results suggest that HIV protease inhibitors tipranavir, indinavir, and saquinavir may bind to EGFR and ERBB3/HER3 interface. Tipranavir and indinavir may also bind to EGFR and ERBB2/HER2 interface. Additionally, a drug used in Alzheimer's disease can bind to RAF1 and BRAF interface. Hence, we propose a methodology to find drugs to be potentially used for cancer using a dataset of structurally similar protein-protein interface clusters rather than pockets in a systematic way.
Collapse
Affiliation(s)
- Ahenk Zeynep Sayin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Rumeli Feneri Yolu Sariyer, 34450, Istanbul, Turkey
| | - Zeynep Abali
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Simge Senyuz
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Fatma Cankara
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, 34450, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Rumeli Feneri Yolu Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
40
|
Huang Q, Mitsiades I, Dowst H, Zarrin-Khameh N, Noor AB, Castro P, Scheurer ME, Godoy G, Mims MP, Mitsiades N. Incidental detection of FGFR3 fusion via liquid biopsy leading to earlier diagnosis of urothelial carcinoma. NPJ Precis Oncol 2023; 7:123. [PMID: 37980380 PMCID: PMC10657397 DOI: 10.1038/s41698-023-00467-9] [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: 03/15/2023] [Accepted: 10/13/2023] [Indexed: 11/20/2023] Open
Abstract
The rising utilization of circulating tumor DNA (ctDNA) assays in Precision Oncology may incidentally detect genetic material from secondary sources. It is important that such findings are recognized and properly leveraged for both diagnosis and monitoring of response to treatment. Here, we report a patient in whom serial cell-free DNA (cfDNA) monitoring for his known prostate adenocarcinoma uncovered the emergence of an unexpected FGFR3-TACC3 gene fusion, a BRCA1 frameshift mutation, and other molecular abnormalities. Due to the rarity of FGFR3 fusions in prostate cancer, a workup for a second primary cancer was performed, leading to the diagnosis of an otherwise-asymptomatic urothelial carcinoma (UC). Once UC-directed treatment was initiated, the presence of these genetic abnormalities in cfDNA allowed for disease monitoring and early detection of resistance, well before radiographic progression. These findings also uncovered opportunities for targeted therapies against FGFR and BRCA1. Overall, this report highlights the multifaceted utility of longitudinal ctDNA monitoring in early cancer diagnosis, disease prognostication, therapeutic target identification, monitoring of treatment response, and early detection of emergence of resistance.
Collapse
Affiliation(s)
- Quillan Huang
- Dept. of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
- Ben Taub General Hospital, Harris Health System, Houston, TX, 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
| | - Irene Mitsiades
- Harvard Medical School, Boston, MA, 02115, USA
- Boston University School of Arts and Sciences, Boston, MA, 02215, USA
| | - Heidi Dowst
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
| | - Neda Zarrin-Khameh
- Ben Taub General Hospital, Harris Health System, Houston, TX, 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
- Dept. of Pathology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Attiya Batool Noor
- Ben Taub General Hospital, Harris Health System, Houston, TX, 77030, USA
| | - Patricia Castro
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
- Dept. of Pathology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michael E Scheurer
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
- Dept. of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Guilherme Godoy
- Ben Taub General Hospital, Harris Health System, Houston, TX, 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
- Dept. of Urology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Martha P Mims
- Dept. of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
- Ben Taub General Hospital, Harris Health System, Houston, TX, 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, 77030, USA
| | - Nicholas Mitsiades
- Department of Internal Medicine, UC Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
41
|
Hatano Y. The Pathology according to p53 Pathway. Pathobiology 2023; 91:230-243. [PMID: 37963443 PMCID: PMC11313058 DOI: 10.1159/000535203] [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: 08/05/2023] [Accepted: 11/12/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Observations play a pivotal role in the progress of science, including in pathology. The cause of a disease such as cancer is analyzed by breaking it down into smaller organs, tissues, cells, and molecules. The current standard cancer diagnostic procedure, microscopic observation, relies on preserved morphological characteristics. In contrast, molecular analyses explore oncogenic pathway activation that leads to genetic mutations and aberrant protein expression. Such molecular analyses could potentially identify therapeutic targets and has gained considerable attention in clinical oncology. SUMMARY This review summarizes the cardinal biomarkers of the p53 pathway, p53, p16, and mouse double minute 2 (MDM2), in the context of traditional surgical pathology and emerging genomic oncology. The p53 pathway, which is dysregulated in more than a half of all cancers, can be applied in several diagnostic settings. A four-classification model of immunophenotype for p53 pathway gene status, tumor types with a high frequency of abnormalities for each p53 pathway gene, and a minimal p53 pathway immunohistochemical panel is also described. KEY MESSAGES Immunohistochemistry of oncogenic signals should be interpreted according to molecular findings based on genomic oncology, in addition to the microscopic findings of diagnostic pathology.
Collapse
Affiliation(s)
- Yuichiro Hatano
- Department of Pathology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| |
Collapse
|
42
|
Chevalier A, Guo T, Gurevich NQ, Xu J, Yajima M, Campbell JD. Characterization of highly active mutational signatures in tumors from a large Chinese population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23297964. [PMID: 37961450 PMCID: PMC10635259 DOI: 10.1101/2023.11.03.23297964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The majority of mutational signatures have been characterized in tumors from Western countries and the degree to which mutational signatures are similar or different in Eastern populations has not been fully explored. We leveraged a large-scale clinical sequencing cohort of tumors from a Chinese population containing 25 tumor types and found that the highly active mutational signatures were similar to those previously characterized1,2. The aristolochic acid signature SBS22 was observed in four soft tissue sarcomas and the POLE-associated signature SBS10 was observed in a gallbladder carcinoma. In lung adenocarcinoma, the polycyclic aromatic hydrocarbon (PAH) signature SBS4 was significantly higher in males compared to females but not associated with smoking status. The UV-associated signature SBS7 was significantly lower in cutaneous melanomas from the Chinese population compared to a similar American cohort. Overall, these results add to our understanding of the mutational processes that contribute to tumors from the Chinese population.
Collapse
Affiliation(s)
- Aaron Chevalier
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Bioinformatics Program, Boston University, Boston, Massachusetts
| | - Tao Guo
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Mathematics & Statistics, Boston University, Boston, Massachusetts
| | - Natasha Q. Gurevich
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Bioinformatics Program, Boston University, Boston, Massachusetts
| | - Jingwen Xu
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Mathematics & Statistics, Boston University, Boston, Massachusetts
| | - Masanao Yajima
- Department of Mathematics & Statistics, Boston University, Boston, Massachusetts
| | - Joshua D. Campbell
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Bioinformatics Program, Boston University, Boston, Massachusetts
| |
Collapse
|
43
|
Lu X, Tawanaie Pour Sedehi N, Su X, Yan F, Alhalabi O, Tannir NM, Malouf GG. Racial Disparities in MiT Family Translocation Renal Cell Carcinoma. Oncologist 2023; 28:1009-1013. [PMID: 37315151 PMCID: PMC10628562 DOI: 10.1093/oncolo/oyad173] [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: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 06/16/2023] Open
Abstract
Racial disparities have been documented in the biology and outcome of certain renal cell carcinomas (RCCs) among Black patients. However, little is known about racial differences in MiT family translocation RCC (TRCC). To investigate this issue, we performed a case-control study using data from The Cancer Genome Atlas (TCGA) and the Chinese OrigiMed2020 cohort. A total of 676 patients with RCC (14 Asian, 113 Black, and 525 White) were identified in TCGA, and TRCC was defined as RCC with TFE3/TFEB translocation or TFEB amplification, leading to 21 patients with TRCC (2 Asian, 8 Black, 10 White, and 1 unknown). Asian (2 of 14 [14.3%] vs 10 of 525 [1.9%]; P = .036) and Black (8 of 113 [7.1%] vs 1.9%; P = .007) patients with RCC showed significantly higher prevalence of TRCC compared with White patients with RCC. The overall mortality rate of TRCC was slightly higher in Asian and Black patients compared with White patients (HR: 6.05, P = .069). OrigiMed2020 Chinese patients with RCC had a significantly higher proportion of TRCC with TFE3 fusions than TCGA White patients with RCC (13 of 250 [5.2%] vs 7 of 525 [1.3%]; P = .003). Black patients with TRCC were more likely to exhibit the proliferative subtype than White patients (6 of 8 [75%] vs 2 of 9 [22.2%]; P = .057) for those who had RNA-seq profiles. We present evidence of higher prevalence of TRCC in Asian and Black patients with RCC compared with White patients and show that these tumors in Asian and Black patients have distinct transcriptional signatures and are associated with poor outcomes.
Collapse
Affiliation(s)
- Xiaofan Lu
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
| | - Nassim Tawanaie Pour Sedehi
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel G Malouf
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
- Department of Medical Oncology, Institut de Cancérologie de Strasbourg, Strasbourg University, Strasbourg, France
| |
Collapse
|
44
|
Wang W, Zhang F, Li Y, Chen B, Gu Y, Shan Y, Li Y, Chen W, Jin Y, Pan L. Whole exome sequencing identifies common mutational landscape of cervix and endometrium small cell neuroendocrine carcinoma. Front Oncol 2023; 13:1182029. [PMID: 37920164 PMCID: PMC10618670 DOI: 10.3389/fonc.2023.1182029] [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: 03/08/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023] Open
Abstract
Background Primary small cell neuroendocrine carcinomas of the cervix and endometrium are rare gynecological malignancies with limited treatment options. This study aimed to improve the understanding of the carcinogenesis process and identify potential therapeutic targets for these two tumor types by constructing the mutational landscape at the whole exome level. Methods Primary tumor tissues and their matched blood samples were obtained from 10 patients with small cell cervical neuroendocrine carcinoma (NECC) and five patients with small cell endometrial neuroendocrine carcinoma (NECE). Whole exome sequencing was performed to construct the somatic mutation profiles. Mutational signature and recurrent mutated gene analysis were used to identify tumor subtypes and common carcinogenesis processes. Results Based on the burden of different mutational signatures, the NECCs in this work can be divided into two subtypes, including the mismatch repair deficiency like (dMMR-like) type (4/10) and the high spontaneous deamination type (6/10). Components of the PI3K/AKT signaling and RAS signaling were exclusively mutated in these two subtypes, respectively. The integration of human papillomavirus made a limited contribution to tumorigenesis in NECC (20%). The dysfunction of the mismatch repair system and microsatellite instability are the major features of NECE. PI3K/AKT, JAK/STAT signaling, and chromatin remodeling activity were the common mutated pathways in NECE. PIK3CA, WNK2, and KMT2B underwent mutations in both the dMMR-like subtype of NECC (50% - 75%) and in NECE (60% - 80%) specimens, while exhibiting infrequent mutational occurrences in publicly available data pertaining to neuroendocrine carcinomas of the lung or bladder (< 10%). Conclusion We identified the two subtypes of NECC with distinct mutated pathways and potential therapy targets. The dMMR-like type NECC and NECE may share a similar carcinogenesis process that include dysfunction of PI3K/AKT signaling, cell cycle, antiapoptotic processes, and chromatin remodeling activity.
Collapse
Affiliation(s)
- Wei Wang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
- Department of Obstetrics and Gynecology, The Fifth People’s Hospital of Ningxia, Shizuishan, China
| | - Fan Zhang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Engineering Medicine, Beihang University, Beijing, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| | - Bo Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| | - Ying Shan
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| | - Yaping Li
- Department of Obstetrics and Gynecology, The Fifth People’s Hospital of Ningxia, Shizuishan, China
| | - Wei Chen
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Engineering Medicine, Beihang University, Beijing, China
| | - Ying Jin
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| | - Lingya Pan
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| |
Collapse
|
45
|
Wang K, Li Z, Xuan Y, Zhao Y, Deng C, Wang M, Xie C, Yuan F, Pang Q, Mao W, Cai D, Zhong Z, Mei J. Pan-cancer analysis of NFE2L2 mutations identifies a subset of lung cancers with distinct genomic and improved immunotherapy outcomes. Cancer Cell Int 2023; 23:229. [PMID: 37794491 PMCID: PMC10552358 DOI: 10.1186/s12935-023-03056-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Mutations in the KEAP1-NFE2L2 signaling pathway were linked to increased tumorigenesis and aggressiveness. Interestingly, not all hotspot mutations on NFE2L2 were damaging; some even were activating. However, there was conflicting evidence about the association between NFE2L2 mutation and Nrf2-activating mutation and responsiveness to immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) and other multiple cancers. METHODS The study with the largest sample size (n = 49,533) explored the landscape of NFE2L2 mutations and their impact response/resistance to ICIs using public cohorts. In addition, the in-house WXPH cohort was used to validate the efficacy of immunotherapy in the NFE2L2 mutated patients with NSCLC. RESULTS In two pan-cancer cohorts, Nrf2-activating mutation was associated with higher TMB value compared to wild-type. We identified a significant association between Nrf2-activating mutation and shorter overall survival in pan-cancer patients and NSCLC patients but not in those undergoing ICIs treatment. Similar findings were obtained in cancer patients carrying the NFE2L2 mutation. Furthermore, in NSCLC and other cancer cohorts, patients with NFE2L2 mutation demonstrated more objective responses to ICIs than patients with wild type. Our in-house WXPH cohort further confirmed the efficacy of immunotherapy in the NFE2L2 mutated patients with NSCLC. Lastly, decreased inflammatory signaling pathways and immune-depleted immunological microenvironments were enriched in Nrf2-activating mutation patients with NSCLC. CONCLUSIONS Our study found that patients with Nrf2-activating mutation had improved immunotherapy outcomes than patients with wild type in NSCLC and other tumor cohorts, implying that Nrf2-activating mutation defined a distinct subset of pan-cancers and might have implications as a biomarker for guiding ICI treatment, especially NSCLC.
Collapse
Affiliation(s)
- Kewei Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Zixi Li
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ying Xuan
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yong Zhao
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chao Deng
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Meidan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chenjun Xie
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Fenglai Yuan
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Qingfeng Pang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Wenjun Mao
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023, China.
| | - Dongyan Cai
- Department of Oncology, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214122, China.
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China.
| | - Jie Mei
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023, China.
| |
Collapse
|
46
|
Bai K, Chen X, Qi X, Zhang Y, Zou Y, Li J, Yu L, Li Y, Jiang J, Yang Y, Liu Y, Feng S, Bu H. Cerebrospinal fluid circulating tumour DNA genotyping and survival analysis in lung adenocarcinoma with leptomeningeal metastases. J Neurooncol 2023; 165:149-160. [PMID: 37897649 PMCID: PMC10638181 DOI: 10.1007/s11060-023-04471-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/26/2023] [Indexed: 10/30/2023]
Abstract
PURPOSE The prognosis of patients with leptomeningeal metastasis (LM) remains poor. Circulating tumour DNA (ctDNA) has been proven to be abundantly present in cerebrospinal fluid (CSF); hence, its clinical implication as a biomarker needs to be further verified. METHODS We conducted a retrospective study of 35 lung adenocarcinoma (LUAD) patients with LM, and matched CSF and plasma samples were collected from all patients. All paired samples underwent next-generation sequencing (NGS) of 139 lung cancer-associated genes. The clinical characteristics and genetic profiling of LM were analysed in association with survival prognosis. RESULTS LM showed genetic heterogeneity, in which CSF had a higher detection rate of ctDNA (P = 0.003), a higher median mutation count (P < 0.0001), a higher frequency of driver mutations (P < 0.01), and more copy number variation (CNV) alterations (P < 0.001) than plasma. The mutation frequencies of the EGFR, TP53, CDKN2A, MYC and CDKN2B genes were easier to detect in CSF than in LUAD tissue (P < 0.05), possibly reflecting the underlying mechanism of LM metastasis. CSF ctDNA is helpful for analysing the mechanism of EGFR-TKI resistance. In cohort 1, which comprised patients who received 1/2 EGFR-TKIs before the diagnosis of LM, TP53 and CDKN2A were the most common EGFR-independent resistant mutations. In cohort 2, comprising those who progressed after osimertinib and developed LM, 7 patients (43.75%) had EGFR CNV detected in CSF but not plasma. Furthermore, patient characteristics and various genes were included for interactive survival analysis. Patients with EGFR-mutated LUAD (P = 0.042) had a higher median OS, and CSF ctDNA mutation with TERT (P = 0.013) indicated a lower median OS. Last, we reported an LM case in which CSF ctDNA dynamic changes were well correlated with clinical treatment. CONCLUSIONS CSF ctDNA could provide a more comprehensive genetic landscape of LM, indicating the potential metastasis-related and EGFR-TKI resistance mechanisms of LM patients. In addition, genotyping of CSF combined with clinical outcomes can predict the prognosis of LUAD patients with LM.
Collapse
Affiliation(s)
- Kaixuan Bai
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Xin Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- Department of Neurology, Xingtai People's Hospital, Xingtai, China
| | - Xuejiao Qi
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yu Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yueli Zou
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Jian Li
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- Department of General Practice, Hengshui People's Hospital, Hengshui, China
| | - Lili Yu
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yuanyuan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Jiajia Jiang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yi Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yajing Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Shuanghao Feng
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Hui Bu
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, China.
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China.
- Neurological Laboratory of Hebei Province, Shijiazhuang, China.
| |
Collapse
|
47
|
Boeschen M, Kuhn CK, Wirtz H, Seyfarth HJ, Frille A, Lordick F, Hacker UT, Obeck U, Stiller M, Bläker H, von Laffert M. Comparative bioinformatic analysis of KRAS, STK11 and KEAP1 (co-)mutations in non-small cell lung cancer with a special focus on KRAS G12C. Lung Cancer 2023; 184:107361. [PMID: 37699269 DOI: 10.1016/j.lungcan.2023.107361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVES Mutations in STK11 (STK11MUT) and KEAP1 (KEAP1MUT) occur frequently in non-small cell lung cancer (NSCLC) and are often co-mutated with KRAS. Several studies linked the co-occurrence of KRASMUT + STK11MUT, as well as KRASMUT + KEAP1MUT to reduced response to immune checkpoint inhibitors (ICI) and even a negative impact on survival. Data focusing STK11 + KEAP1 co-mutations or the triple mutation (KRAS + STK11 + KEAP1) are scarce. The recent availability of KRAS-G12C inhibitors increases the clinical relevance of those co-mutations in KRAS-mutated NSCLC. MATERIALS AND METHODS We present a comprehensive bioinformatic analysis encompassing six datasets retrieved from cBioPortal. RESULTS Independent of the treatment, triple mutations and STK11MUT + KEAP1MUT were significantly associated with a reduced overall survival (OS). Across treatments, OS of patients with a KRAS G12C triple mutation was significantly reduced compared to patients with KRAS G12C-only. Under ICI-therapy, there was no significant difference in OS between patients harboring the KRAS G12C-only and patients with the KRAS G12C triple mutation, but a significant difference between patients harboring KRAS non-G12C and KRAS non-G12C triple mutations. Triple mutated primary tumors showed a significantly increased frequency of distant metastases to bone and adrenal glands compared to KRAS-only mutated tumors. Additionally, our drug response analysis in cancer cell lines harboring the triple mutations revealed the WNT pathway inhibitor XAV-939 as a potential future drug candidate for this mutational situation. CONCLUSION The triple mutation status may serve as a negative prognostic and predictive factor across treatments compared to KRASMUT-only. KRAS G12C generally seems to be a negative predictive marker for ICI-therapy.
Collapse
Affiliation(s)
- Myriam Boeschen
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany.
| | - Christina Katharina Kuhn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Hubert Wirtz
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Hans-Jürgen Seyfarth
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Armin Frille
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Florian Lordick
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ulrich T Hacker
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ulrike Obeck
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Mathias Stiller
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Hendrik Bläker
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Maximilian von Laffert
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany.
| |
Collapse
|
48
|
Mansur MB, Greaves M. Convergent TP53 loss and evolvability in cancer. BMC Ecol Evol 2023; 23:54. [PMID: 37743495 PMCID: PMC10518978 DOI: 10.1186/s12862-023-02146-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/10/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer cell populations evolve by a stepwise process involving natural selection of the fittest variants within a tissue ecosystem context and as modified by therapy. Genomic scrutiny of patient samples reveals an extraordinary diversity of mutational profiles both between patients with similar cancers and within the cancer cell population of individual patients. Does this signify highly divergent evolutionary trajectories or are there repetitive and predictable patterns?Major evolutionary innovations or adaptations in different species are frequently repeated, or convergent, reflecting both common selective pressures and constraints on optimal solutions. We argue this is true of evolving cancer cells, especially with respect to the TP53 gene. Functional loss variants in TP53 are the most common genetic change in cancer. We discuss the likely microenvironmental selective pressures involved and the profound impact this has on cell fitness, evolvability and probability of subsequent drug resistance.
Collapse
Affiliation(s)
- Marcela Braga Mansur
- Centre for Evolution and Cancer, The Institute of Cancer Research, ICR, London, UK
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, ICR, London, UK.
| |
Collapse
|
49
|
Gandhi MM, Ricciuti B, Harada G, Repetto M, Gildenberg MS, Singh A, Li YY, Gagné A, Wang X, Aizer A, Fitzgerald K, Nishino M, Alessi J, Pecci F, Di Federico A, Fisch A, Drilon A, Nardi V, Sholl L, Awad MM, Rotow J. Amplification of Wild-Type RET Represents a Novel Molecular Subtype of Several Cancer Types With Clinical Response to Selpercatinib. JCO Precis Oncol 2023; 7:e2300295. [PMID: 37972337 PMCID: PMC10681403 DOI: 10.1200/po.23.00295] [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: 06/09/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE RET rearrangements and RET activating point mutations represent targetable genomic alterations in advanced solid tumors. However, the frequency and clinicopathologic characteristics of wild-type RET amplification in cancer and its potential role as a targetable oncogenic driver are not well-characterized. METHODS In two institutional cohorts of patients with solid cancers from the Dana-Farber Cancer Institute (DFCI) and Memorial Sloan Kettering Cancer Center (MSKCC) whose tumors underwent next-generation sequencing (NGS), the frequency and clinicopathologic features of wild-type RET amplification in the absence of RET rearrangements or activating mutations was assessed. The findings were validated using merged data from The Cancer Genome Atlas (TCGA), Genomics Evidence Neoplasia Information Exchange (GENIE), and China Pan-Cancer data sets. RESULTS The frequency of wild-type RET amplification across all solid cancers was 0.08% (26 of 32,505) in the DFCI cohort, 0.05% (26 of 53,152) in the MSKCC cohort, and 0.25% (71 of 28,623) in the cohort from TCGA, GENIE, and China Pan-Cancer. Cancer types with RET amplification included non-small-cell lung cancer (NSCLC), hepatobiliary cancer, prostate cancer, breast cancer, and others. The median RET copy number in RET-amplified cases was 7.5 (range, 6-36) in the DFCI cohort and 5.7 (range, 4-27.7) in the MSKCC cohort. Among 11 RET-amplified NSCLCs, eight had no other concurrent driver mutations. Finally, we report on a 69-year-old man with recurrent NSCLC harboring high-level wild-type RET amplification (22-28 copies) as the only identified putative genomic driver who experienced both a systemic and intracranial confirmed response to the RET inhibitor selpercatinib. CONCLUSION Amplification of wild-type RET represents a novel, targetable molecular subset of cancer.
Collapse
Affiliation(s)
- Malini M. Gandhi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Guilherme Harada
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Matteo Repetto
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | | | - Ankit Singh
- Center for Integrated Diagnostics, Massachusetts General Hospital, Boston, MA
| | - Yvonne Y. Li
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Andréanne Gagné
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Xinan Wang
- Harvard School of Public Health, Boston, MA
| | - Ayal Aizer
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kelly Fitzgerald
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA
| | - Joao Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Federica Pecci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Adam Fisch
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Mark M. Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Julia Rotow
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
50
|
Hao S, Zhao X, Fan Y, Liu Z, Zhang X, Li W, Yuan H, Zhang J, Zhang Y, Ma T, Tao H. Prevalence and spectrum of cancer predisposition germline mutations in young patients with the common late-onset cancers. Cancer Med 2023; 12:18394-18404. [PMID: 37610374 PMCID: PMC10524041 DOI: 10.1002/cam4.6445] [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: 02/12/2023] [Revised: 06/30/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Pathogenic germline variants (PGVs) can play a vital role in the oncogenesis process in carriers. Previous studies have recognized that PGVs contribute to early onset of tumorigenesis in certain cancer types, for example, colorectal cancer and breast cancer. However, the reported prevalence data of cancer-associated PGVs were highly inconsistent due to nonuniform patient cohorts, sequencing methods, and prominent difficulties in pathogenicity interpretation of variants. In addition to the above difficulties, due to the rarity of cases, the prevalence of cancer PGV carriers in young cancer patients affected by late-onset cancer types has not been comprehensively evaluated to date. METHODS A total of 131 young cancer patients (1-29 years old at diagnosis) were enrolled in this study. The patients were affected by six common late-onset cancer types, namely, lung cancer, liver cancer, colorectal cancer, gastric cancer, renal cancer, and head-neck cancer. Cancer PGVs were identified and analyzed. based on NGS-based targeted sequencing followed by bioinformatic screening and strict further evaluations of variant pathogenicity. RESULTS Twenty-three cancer PGVs in 21 patients were identified, resulting in an overall PGV prevalence of 16.0% across the six included cancer types, which was approximately double the prevalence reported in a previous pancancer study. Nine of the 23 PGVs are novel, thus expanding the cancer PGV spectrum. Seven of the 23 (30.4%) PGVs are potential therapeutic targets of olaparib, with potential implications for clinical manipulation. Additionally, a small prevalence of somatic mutations of some classic cancer hallmark genes in young patients, in contrast to all-age patients, was revealed. CONCLUSION This study demonstrates the high prevalence of PGVs in young cancer patients with the common late-onset cancers and the potentially significant clinical implications of cancer PGVs, the findings highlight the value of PGV screening in young patients across lung cancer, liver cancer, colorectal cancer, gastric cancer, renal cancer, or head-neck cancer.
Collapse
Affiliation(s)
- Shaoyu Hao
- Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ximeng Zhao
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | - Yue Fan
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhong Shan Hospital, Fudan UniversityShanghaiChina
| | - Zhengchuang Liu
- Key Laboratory of Gastroenterology of Zhejiang ProvinceZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeHangzhouChina
| | - Xiang Zhang
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | - Wei Li
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | | | - Jie Zhang
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | | | - Tonghui Ma
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | - Houquan Tao
- Key Laboratory of Gastroenterology of Zhejiang ProvinceZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeHangzhouChina
- Department of GastroenterologyZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeHangzhouChina
| |
Collapse
|