1
|
Russo G, Scimone C, Palumbo L, Roscigno G, Sarracino C, Tomaiuolo I, Pisapia P, Pepe F, Rocco D, Gridelli C, Troncone G, Malapelle U. Biologics for novel driver altered non-small cell lung cancer: potential and pitfalls. Crit Rev Oncol Hematol 2025; 212:104748. [PMID: 40324663 DOI: 10.1016/j.critrevonc.2025.104748] [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: 03/06/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
Precision medicine has revolutionized clinical paradigm of lung cancer (LC) patients optimizing therapeutical options on the basis of molecular fingerprinting of tumor cells. The advent of the genomic era contributed to the widespread diffusion of sequencing technologies laying the basis for the approval of an increasing number of clinically relevant predictive biomarkers in clinical settings. In the rapidly evolving scenario of predictive biomarkers, mandatory testing genes demonstrated a statistically significant clinical benefit in LC patients elected to molecular tests, but emerging biomarkers are under investigation to raise the bar in the clinical management of LC patients. To date, promising IHC-based predictive biomarkers emerged as potentially integrative tools in the panel of clinically approved biomarkers. On this basis, genomic, transcriptomic and proteomic data are gaining ground toward "3D" biology" supporting the need of a multidimensional analysis of tumor cells to clinically stratify LC patients. Here we sought to overview the most promising biomarkers investigated in clinical trials to be integrated into diagnostic panel of predictive biomarkers tools for NSCLC patients.
Collapse
Affiliation(s)
- Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Claudia Sarracino
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Ilaria Tomaiuolo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Napoli, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Avellino, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy.
| |
Collapse
|
2
|
Shen Y, Chen JQ, Li XP. Differences between lung adenocarcinoma and lung squamous cell carcinoma: Driver genes, therapeutic targets, and clinical efficacy. Genes Dis 2025; 12:101374. [PMID: 40083325 PMCID: PMC11904499 DOI: 10.1016/j.gendis.2024.101374] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/15/2024] [Accepted: 06/22/2024] [Indexed: 03/16/2025] Open
Abstract
With the rapid advancements in second-generation gene sequencing technologies, a growing number of driver genes and associated therapeutic targets have been unveiled for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). While they are clinically classified as non-small cell lung cancer (NSCLC), they display distinct genomic features and substantial variations in clinical efficacy, underscoring the need for particular attention. Hence, this review provides a comprehensive overview of the latest advancements in driver genes, epigenetic targets, chemotherapy, targeted therapy, and immunotherapy for LUAD and LUSC. Additionally, it delves into the distinctions in signaling pathways and pivotal facets of clinical management specific to these two categories of lung cancer. Moreover, we furnish pertinent details regarding clinical trials pertaining to driver genes and epigenetics, thus establishing a theoretical foundation for the realization of precision treatments for LUAD and LUSC.
Collapse
Affiliation(s)
- Yue Shen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jie-Qi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang-Ping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
3
|
Luo W, Xu M, Wong N, Ng CSH. Alternative Splicing in Lung Adenocarcinoma: From Bench to Bedside. Cancers (Basel) 2025; 17:1329. [PMID: 40282505 PMCID: PMC12025742 DOI: 10.3390/cancers17081329] [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: 01/25/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a highly heterogeneous tumor and the most prevalent pathological type of lung cancer. The alternative splicing (AS) of mRNA enables the generation of multiple protein products from a single gene. This is a tightly regulated process that significantly contributes to the proteome diversity in eukaryotes. Recent multi-omics studies have delineated the splicing profiles that underline LUAD tumorigenesis from initiation to metastasis. Such progress holds robust promise to facilitate the development of screening strategies and individualized therapies. Perturbed AS fosters the emergence of novel neoantigen resources and disturbances in the immune microenvironment, which allow new investigations into modulatory targets for LUAD immunotherapy. This review presents an update on the landscape of dysregulated splicing events in LUAD and the associated mechanisms and theranostic perspectives with unique insights into AS-based immunotherapy, such as Chimeric Antigen Receptor T cell therapy. These AS variants can be used in conjunction with current therapeutic modules in LUAD, allowing bench to bedside translation to combat this highly malignant cancer.
Collapse
Affiliation(s)
| | | | - Nathalie Wong
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; (W.L.); (M.X.)
| | - Calvin Sze-Hang Ng
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; (W.L.); (M.X.)
| |
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
|
Chung C, Umoru G. Prognostic and predictive biomarkers with therapeutic targets in nonsmall-cell lung cancer: A 2023 update on current development, evidence, and recommendation. J Oncol Pharm Pract 2025; 31:438-461. [PMID: 38576390 DOI: 10.1177/10781552241242684] [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] [Indexed: 04/06/2024]
Abstract
BackgroundSince the publication of the original work in 2014, significant progress has been made in the characterization of genomic alterations that drive oncogenic addiction of nonsmall cell lung cancer (NSCLC) and how the immune system can leverage non-oncogenic pathways to modulate therapeutic outcomes. This update evaluates and validates the recent and emerging data for prognostic and predictive biomarkers with therapeutic targets in NSCLC.Data sourcesWe performed a literature search from January 2015 to October 2023 using the keywords non-small cell lung cancer, clinical practice guidelines, gene mutations, genomic assay, immune cancer therapy, circulating tumor DNA, predictive and prognostic biomarkers, and targeted therapies.Study selection and data extractionWe identified, reviewed, and evaluated relevant clinical trials, meta-analyses, seminal articles, and published clinical practice guidelines in the English language.Data synthesisRegulatory-approved targeted therapies include those somatic gene alterations of EGFR ("classic" mutations, exon 20 insertion, and rare EGFR mutations), ALK, ROS1, BRAF V600, RET, MET, NTRK, HER2, and KRAS G12C. Data for immunotherapy and circulating tumor DNA in next-generation sequencing are considered emerging, whereas the predictive role for PIK3CA gene mutation is insufficient.ConclusionsAdvances in sequencing and other genomic technologies have led to identifying novel oncogenic drivers, novel resistance mechanisms, and co-occurring mutations that characterize NSCLC, creating further therapeutic opportunities. The benefits associated with immunotherapy in the perioperative setting hold initial promise, with their long-term results awaiting.
Collapse
Affiliation(s)
- Clement Chung
- Department of Pharmacy, Houston Methodist West Hospital, Houston, TX, USA
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
6
|
Galeș LN, Păun MA, Butnariu I, Simion L, Manolescu LSC, Trifănescu OG, Anghel RM. Next-Generation Sequencing in Oncology-A Guiding Compass for Targeted Therapy and Emerging Applications. Int J Mol Sci 2025; 26:3123. [PMID: 40243903 PMCID: PMC11988731 DOI: 10.3390/ijms26073123] [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: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Multigene sequencing technologies provide a foundation for targeted therapy and precision oncology by identifying actionable alterations and enabling the development of treatments that substantially improve clinical outcomes. This review emphasizes the importance of having a molecular compass guiding treatment decision-making through the multitude of alterations and genetic mutations, showcasing why NGS plays a pivotal role in modern oncology.
Collapse
Affiliation(s)
- Laurenția Nicoleta Galeș
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Medical Oncology II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai-Andrei Păun
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
| | - Ioana Butnariu
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Laurentiu Simion
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Loredana Sabina Cornelia Manolescu
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Clinical Laboratory of Medical Microbiology, “Marius Nasta” Institute of Pneumology, 050159 Bucharest, Romania
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Oana Gabriela Trifănescu
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
7
|
Sattler M, Salgia R. The expanding role of the receptor tyrosine kinase MET as a therapeutic target in non-small cell lung cancer. Cell Rep Med 2025; 6:101983. [PMID: 40020676 PMCID: PMC11970332 DOI: 10.1016/j.xcrm.2025.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Aberrant regulation of MET receptor tyrosine kinase activity is a frequent event in non-small cell lung cancer (NSCLC), even though the frequency of oncogenic driver mutations of MET is low. Our discovery of oncogenic MET exon 14 skipping mutations, the characterization of the first prototype MET kinase inhibitor, and characterization of MET expression levels have led the way to novel therapeutic approaches with improved outcomes in NSCLC. MET exon 14 mutations are the most consequential but not the only alterations that can be targeted through small molecule tyrosine kinase inhibitors. The abundant expression of cellular MET (c-MET) in cancer cells has provided new opportunities for immuno-oncology approaches in a broader patient population, and the integration of MET-targeted personalized medicine with immunotherapy has not been fully exploited yet. Here, we highlight essential facets of MET as a therapeutic target in NSCLC and provide an outlook for future approaches.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
8
|
Bouchard N, Daaboul N. Lung Cancer: Targeted Therapy in 2025. Curr Oncol 2025; 32:146. [PMID: 40136350 PMCID: PMC11941068 DOI: 10.3390/curroncol32030146] [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: 01/31/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Lung cancer treatment has changed in the last twenty years since the discovery of EGFR mutations. In this article, we will review the current state of the art for non-small cell lung cancer (NSCLC) actionable genomic alterations (AGA). AGAs are mostly found in lung adenocarcinomas, a subtype of non-small cell lung cancers. We will focus on the current treatment for EGFR mutations, ALK fusions, ROS1 fusions, BRAF V600E mutations, MET exon 14-skipping mutations, RET fusions, KRAS G12C mutations, ERBB2 mutations (also called HER2 mutations), and NTRK fusions. We will also touch on the key toxicities associated with these medications. Treatments are mostly available for the metastatic stage, but we will also discuss adjuvant therapy for EGFR mutations and ALK fusions, as well as stage III post-chemoradiotherapy treatment for EGFR lung cancer.
Collapse
Affiliation(s)
- Nicole Bouchard
- Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Nathalie Daaboul
- Centre Intégré de Cancérologie de la Montérégie, Université de Sherbrooke, Longueuil, QC J4V 2H1, Canada
| |
Collapse
|
9
|
Morin C, Mazières J. [Lung cancers with rare oncogenic drivers: RET, ROS-1, MET, HER2 and BRAF]. Bull Cancer 2025; 112:3S117-3S126. [PMID: 40155071 DOI: 10.1016/s0007-4551(25)00165-1] [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] [Indexed: 04/01/2025]
Abstract
In non-small cell lung cancer, the presence of an oncogenic driver is frequently documented. Advances in molecular biology have enabled the identification of so-called rare oncogenic addictions, with an incidence of less than 5%, such as ROS-1 and RET rearrangements, and MET, BRAF and HER2 mutations. Targeted therapies have shown strong tumor responses with a better tolerance profile compared to chemotherapy. Consequently, targeted therapies have revolutionized the therapeutic landscape, particularly as immune checkpoint inhibitors are often ineffective in the presence of an oncogenic driver. To ensure optimal management in the era of personalized medicine, it is recommended to screen for oncogenic addictions, including rare ones, at diagnosis. In this review, we discuss the targeted therapies available in France and the promising future molecules for managing rare oncogenic drivers. Targeted therapies have already proven their efficacy as first-line treatments for ROS-1 and RET alterations, and as second-line treatments for MET and BRAF mutations.
Collapse
Affiliation(s)
- Clara Morin
- Service de pneumologie, hôpital Larrey, CHU de Toulouse, Toulouse, France; Université Paul-Sabatier, Toulouse, France; Centre de recherche de cancérologie de Toulouse (CRCT), Inserm, Toulouse, France.
| | - Julien Mazières
- Service de pneumologie, hôpital Larrey, CHU de Toulouse, Toulouse, France; Université Paul-Sabatier, Toulouse, France; Centre de recherche de cancérologie de Toulouse (CRCT), Inserm, Toulouse, France
| |
Collapse
|
10
|
Guérin C, Vinchent A, Fernandes M, Damour I, Laratte A, Tellier R, Estevam GO, Meneboo JP, Villenet C, Descarpentries C, Fraser JS, Figeac M, Cortot AB, Rouleau E, Tulasne D. MET variants with activating N-lobe mutations identified in hereditary papillary renal cell carcinomas still require ligand stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.03.565283. [PMID: 37965202 PMCID: PMC10635098 DOI: 10.1101/2023.11.03.565283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
In hereditary papillary renal cell carcinoma (HPRCC), the hepatocyte growth factor receptor (MET) receptor tyrosine kinase (RTK) mutations recorded to date are located in the kinase domain and lead to constitutive MET activation. This contrasts with MET mutations identified in non-small cell lung cancer (NSCLC), which lead to exon 14 skipping and deletion of a regulatory domain: in this latter case, the mutated receptor still requires ligand stimulation. Sequencing of MET in samples from 158 HPRCC and 2808 NSCLC patients revealed ten uncharacterized mutations. Four of these, all found in HPRCC and leading to amino acid substitutions in the N-lobe of the MET kinase, proved able to induce cell transformation, which was further enhanced by hepatocyte growth factor (HGF) stimulation: His1086Leu, Ile1102Thr, Leu1130Ser and Cis1125Gly. Similar to the variant resulting in MET exon 14 skipping, the two N-lobe MET variants His1086Leu and Ile1102Thr were found to require stimulation by HGF in order to strongly activate downstream signaling pathways and epithelial cell motility. The Ile1102Thr mutation also displayed transforming potential, promoting tumor growth in a xenograft model. In addition, the N-lobe-mutated MET variants were found to trigger a common HGF-stimulation-dependent transcriptional program, consistent with an observed increase in cell motility and invasion. Altogether, this functional characterization revealed that N-lobe variants still require ligand stimulation, in contrast to other RTK variants. This suggests that HGF expression in the tumor microenvironment is important for tumor growth. The sensitivity of these variants to MET inhibitors opens the way for use of targeted therapies for patients harboring the corresponding mutations.
Collapse
|
11
|
CHEN J, HAN B, HU Y, HU J, Cancer Specialty Committee of Chinese Elderly Health Care Association L. [Expert Consensus on Diagnosis and Treatment of NSCLC with MET Abnormalities
(2025 Version)]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:81-94. [PMID: 40114485 PMCID: PMC11931236 DOI: 10.3779/j.issn.1009-3419.2025.102.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Indexed: 03/22/2025]
Abstract
The mesenchymal-epithelial transition factor (MET) gene, located on human chromosome 7, plays a crucial role in the regulation of physiological processes such as cell proliferation, migration, invasion, and angiogenesis. The MET gene is one of the key drivers in non-small cell lung cancer (NSCLC), with various forms of abnormalities including MET exon 14 (METex14) skipping mutations, MET gene amplification, MET fusions, MET protein overexpression, MET activating mutations and etc. With an increasing understanding of the mechanisms underlying MET abnormalities, therapeutic strategies targeting these abnormalities have gained significant attention, and numerous studies have confirmed that NSCLC patients with MET abnormalities can derive substantial benefits from such treatments. Lung Cancer Specialty Committee of Chinese Elderly Health Care Association organized a panel of experts to provide professional recommendations on current clinical issues in the diagnosis and treatment of MET-aberrant NSCLC, combining clinical practice experiences and evidence-based medical evidences. The "Expert Consensus on Diagnosis and Treatment of NSCLC with MET Abnormalities (2025 Version)" has been formulated to provide standardized guidances for clinical practice in China, with the aim of optimizing the treatment outcomes.
.
Collapse
|
12
|
Guérin C, Vinchent A, Fernandes M, Damour I, Laratte A, Tellier R, Estevam GO, Meneboo JP, Villenet C, Descarpentries C, Fraser JS, Figeac M, Cortot AB, Rouleau E, Tulasne D. MET variants with activating N-lobe mutations identified in hereditary papillary renal cell carcinomas still require ligand stimulation. Mol Oncol 2025. [PMID: 39980226 DOI: 10.1002/1878-0261.13806] [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: 03/14/2024] [Revised: 10/16/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
In hereditary papillary renal cell carcinoma (HPRCC), the hepatocyte growth factor receptor (MET) receptor tyrosine kinase (RTK) mutations recorded to date are located in the kinase domain and lead to constitutive MET activation. This contrasts with MET mutations identified in non-small-cell lung cancer (NSCLC), which lead to exon 14 skipping and deletion of a regulatory domain: In this latter case, the mutated receptor still requires ligand stimulation. Sequencing of MET in samples from 158 HPRCC and 2808 NSCLC patients revealed 10 uncharacterized mutations. Four of these, all found in HPRCC and leading to amino acid substitutions in the N-lobe of the MET kinase, proved able to induce cell transformation, which was further enhanced by hepatocyte growth factor (HGF) stimulation: His1086Leu, Ile1102Thr, Leu1130Ser, and Cis1125Gly. Similar to the variant resulting in MET exon 14 skipping, the two N-lobe MET variants His1086Leu and Ile1102Thr were found to require stimulation by HGF in order to strongly activate downstream signaling pathways and epithelial cell motility. The Ile1102Thr mutation also displayed transforming potential, promoting tumor growth in a xenograft model. In addition, the N-lobe-mutated MET variants were found to trigger a common HGF-stimulation-dependent transcriptional program, consistent with an observed increase in cell motility and invasion. Altogether, this functional characterization revealed that N-lobe variants still require ligand stimulation, in contrast to other RTK variants. This suggests that HGF expression in the tumor microenvironment is important for tumor growth. The sensitivity of these variants to MET inhibitors opens the way for use of targeted therapies for patients harboring the corresponding mutations.
Collapse
Affiliation(s)
- Célia Guérin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Audrey Vinchent
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Marie Fernandes
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Isabelle Damour
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Agathe Laratte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Rémi Tellier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Gabriella O Estevam
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Jean-Pascal Meneboo
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Céline Villenet
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Clotilde Descarpentries
- Department of Biochemistry and Molecular Biology, Hormonology Metabolism Nutrition Oncology, CHU Lille, France
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Martin Figeac
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Alexis B Cortot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
- Thoracic Oncology Department, Univ. Lille, CHU Lille, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, Villejuif, France
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
13
|
Wang M, Zhang S, Yi D, Ou Y, Xie S, Zeng C, Qin X, Zhao L, Wang Z, Kong F, Chen L. Advances in clinical research of MET exon 14 skipping mutations in non-small cell lung cancer. J Cancer Res Clin Oncol 2025; 151:78. [PMID: 39937291 PMCID: PMC11821758 DOI: 10.1007/s00432-025-06115-y] [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: 08/09/2023] [Accepted: 01/21/2025] [Indexed: 02/13/2025]
Abstract
The cellular-mesenchymal to epithelial transition factor (MET) gene plays a crucial role in maintaining cell homeostasis, motility, and apoptosis. In cancer, MET gene alterations promote tumour cell proliferation, invasion and metastasis. In non-small cell lung cancer (NSCLC), MET gene alterations include MET exon 14 (METex14) skipping mutation (METΔ14ex), MET amplification (METamp), MET fusion, and MET tyrosine kinase domain missense mutations (MET-TKD) and MET protein overexpression. Among them, the METΔ14ex is an independent driver gene of NSCLC. Three to four per cent of NSCLC patients carry METΔ14ex, and these patients have a poor prognosis and respond poorly to conventional chemotherapy. Small molecule highly selective MET inhibitors such as carmatinib, tepotinib, and cervotinib have shown promising efficacy and safety in clinical trials. Monoclonal antibodies, bispecific antibodies, antibody conjugate drugs, and immune checkpoint inhibitors provide more treatment space for patients with METΔ14ex. In this review, we summarize the current application and research of MET inhibitors and immune checkpoint inhibitors in NSCLC with METΔ14ex and provide recommendations for precise treatment of NSCLC patients with MET gene changes mutations. It also provides new ideas for solving the problems of synergistic effect and drug resistance in targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Mengchao Wang
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Shao Zhang
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Dan Yi
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Yan Ou
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Shuqi Xie
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Chuanxiu Zeng
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Xueqian Qin
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Lu Zhao
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Zhen Wang
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Fanming Kong
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Liwei Chen
- Oncology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
14
|
Oksen D, Boutmy E, Wang Y, Stroh C, Johne A, Nisbett AR, Ryder A. Patients With Advanced Non-small Cell Lung Cancer Harboring MET Alterations: A Descriptive Cohort Study. Clin Lung Cancer 2025:S1525-7304(25)00024-5. [PMID: 40011191 DOI: 10.1016/j.cllc.2025.01.013] [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: 08/21/2024] [Revised: 12/02/2024] [Accepted: 01/29/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE Mesenchymal-epithelial transition factor (MET) alterations are rare oncogenic drivers in patients with non-small cell lung cancer (NSCLC). This study characterized patients with advanced NSCLC harboring MET exon 14 (METex14) skipping and MET amplification (METamp) in routine clinical practice in the United States. PATIENTS AND METHODS Using electronic medical record data from the ConcertAI Oncology Dataset (2004-2022), 2 patient cohorts were identified: 1 with METex14 skipping with or without METamp, and 1 with METamp without METex14 skipping. A subgroup with METamp and concomitant epidermal growth factor receptor (EGFR) mutations who had received EGFR-tyrosine kinase inhibitors (TKIs) was also analyzed. Patient characteristics, treatment patterns and outcomes were described. RESULTS 93 patients with advanced NSCLC harboring METex14 skipping and 164 with advanced NSCLC harboring METamp were identified. Established oncogenic drivers other than MET were less frequent in the METex14 skipping cohort than the METamp cohort. In both cohorts, first-line chemotherapy use decreased over time while immune checkpoint inhibitor (ICI) and MET inhibitor use increased. Treatment patterns were heterogeneous, with patients receiving multiple drug classes across therapy lines. Outcomes were better for patients with METex14 skipping NSCLC who received targeted therapies or ICIs versus chemotherapy. Subgroup analyses of EGFR-TKI-treated patients with METamp indicated poor outcomes. CONCLUSION Patients with METex14 skipping and/or METamp NSCLC require targeted and personalized treatment approaches to optimize treatment effect and have an unmet medical need. With targeted therapies recently available and others under exploration, treatment outcomes could significantly improve for patients with NSCLC harboring these rare drivers.
Collapse
Affiliation(s)
- Dina Oksen
- Department of Epidemiology, the healthcare business of Merck KGaA, Darmstadt, Germany.
| | - Emmanuelle Boutmy
- Department of Epidemiology, the healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Christopher Stroh
- Department of Epidemiology, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Andreas Johne
- Department of Epidemiology, the healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Alex Ryder
- West Cancer Center and Research Institute, Germantown, TN
| |
Collapse
|
15
|
Li M, Huang J, Xing R, Du X, Wei C, Wang H. Exploring practical experience with different treatments in NSCLC patients with MET-deregulated: a retrospective analysis from the real world. BMC Pulm Med 2025; 25:35. [PMID: 39856706 PMCID: PMC11761713 DOI: 10.1186/s12890-024-03437-4] [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: 01/08/2024] [Accepted: 12/05/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Mesenchymal to epithelial transition factor (MET) dysregulation in non-small-cell-lung-cancer (NSCLC) is understudied, with scant data on treatment outcomes. METHODS We retrospectively examined 160 NSCLC patients: 125 with primary MET mutations (further classified into MET exon 14 (METex14) skipping mutations and primary MET amplifications) and 35 with secondary MET amplifications. Patients underwent varied treatments: Chemotherapy, Immune monotherapy, Crizotinib, or Savolitinib. Secondary MET amplification patients were grouped by treatment: Group A (Class Ib MET-TKI with third-generation EGFR-TKI), Group B (Crizotinib with first-generation EGFR-TKI), and Group C (Crizotinib alone). One hundred and thirty patients have completed the whole treatment process. Their data were included in the study's survival analysis (included 95 patients with primary MET mutations and 35 patients with secondary MET amplifications). RESULTS Among METex14 skipping mutations patients (n = 57), median progression free survival (PFS) was: Chemotherapy 7.64 m, Crizotinib 8.5 m, Savolitinib 9.3 m, and Immunotherapy 3.87 m. Targeted therapies and chemotherapy significantly outperformed Immunotherapy. Sub-group analysis indicated splice donor region mutations benefited more than those at the polypyrimidine tract (9.23 m vs. 4.03 m, P = 0.038). For primary MET amplifications (n = 38), PFS was: Chemotherapy 2.84 m, Crizotinib 3.80 m, Savolitinib 5.23 m, and Immunotherapy 3.30 m. Patients with copy number (CN) > 5 had longer PFS than CN ≤ 5 (5.17 m vs. 3.44 m, P = 0.039). In secondary MET amplifications (n = 35), Group A and B had similar PFS (6.77 m and 6.57 m) versus Group C (3.13 m). Dual-target therapy PFS showed no difference between CN ≤ 5 and CN > 5 (8.63 m vs. 6.27 m, P = 0.29). CONCLUSION NSCLC patients with METex14 skipping mutations benefit more from targeted therapies, especially those with splice donor mutations. MET amplification patients benefit universally from targeted therapies; primary MET amplifications show higher benefits with increased copy numbers. For secondary MET amplifications post-EGFR-TKI resistance, dual-target therapy surpasses Crizotinib monotherapy, independent of MET copy number.
Collapse
Affiliation(s)
- Mengmeng Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Jiuyan Huang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Ruyue Xing
- Department of Medical Oncology, Nanyang Central Hospital, Nanyang, 473000, Henan, China
| | - Xinyang Du
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Chunhua Wei
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Huijuan Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China.
| |
Collapse
|
16
|
Okun SA, Lu D, Sew K, Subramaniam A, Lockwood WW. MET Activation in Lung Cancer and Response to Targeted Therapies. Cancers (Basel) 2025; 17:281. [PMID: 39858062 PMCID: PMC11764361 DOI: 10.3390/cancers17020281] [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: 12/11/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor receptor (MET) is a receptor tyrosine kinase (RTK) that mediates the activity of a variety of downstream pathways upon its activation. These pathways regulate various physiological processes within the cell, including growth, survival, proliferation, and motility. Under normal physiological conditions, this allows MET to regulate various development and regenerative processes; however, mutations resulting in aberrant MET activity and the consequent dysregulation of downstream signaling can contribute to cellular pathophysiology. Mutations within MET have been identified in a variety of cancers and have been shown to mediate tumorigenesis by increasing RTK activity and downstream signaling. In lung cancer specifically, a number of patients have been identified as possessing MET alterations, commonly receptor amplification (METamp) or splice site mutations resulting in loss of exon 14 (METex14). Due to MET's role in mediating oncogenesis, it has become an attractive clinical target and has led to the development of various targeted therapies, including MET tyrosine kinase inhibitors (TKIs). Unfortunately, these TKIs have demonstrated limited clinical efficacy, as patients often present with either primary or acquired resistance to these therapies. Mechanisms of resistance vary but often occur through off-target or bypass mechanisms that render downstream signaling pathways insensitive to MET inhibition. This review provides an overview of the therapeutic landscape for MET-positive cancers and explores the various mechanisms that contribute to therapeutic resistance in these cases.
Collapse
Affiliation(s)
- Sarah Anna Okun
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Daniel Lu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Katherine Sew
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Asha Subramaniam
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Department of Pathology and Laboratory Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - William W. Lockwood
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Pathology and Laboratory Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
17
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [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: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Shields MD, Minton KG, Tran M, Gunderman PR, Larsson LG, Guo S, Kniese CM, Wei CX, Marin-Acevedo JA, Maniar R, Durm GA, He W, Hanna NH. Defining the needle in a haystack: A compendium of genomic, pathologic, and clinical characteristics of rare pulmonary tumors. Lung Cancer 2025; 199:108035. [PMID: 39731864 DOI: 10.1016/j.lungcan.2024.108035] [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: 09/06/2024] [Revised: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 12/30/2024]
Abstract
A major paradigm shift in the diagnosis, management, and survival outcomes of early and advanced non-small cell lung cancer has transpired over the past few decades in thoracic oncology with the incorporation of molecular testing, targeted therapy, immunotherapy, neoadjuvant, and adjuvant approaches. However, transformation in the management and survival outcomes of rare lung tumors is lacking. Given the scarcity of these tumor types, randomized trials are rarely performed, and treatment is extrapolated from case series, tumor-agnostic trials, or cancers with similar histology. Literature informing the management of rare pulmonary tumors is typically limited to a single histology, unique features, or extraordinary responses to therapy. Few resources detailing genomic characteristics and delineating features of these tumors are available, often resulting in suboptimal treatment. Here, we explore the clinical, histopathologic, genomic features and potential therapies of five rare pulmonary tumors, namely adenosquamous, basaloid squamous, mucoepidermoid, carcinosarcoma, and NUT carcinoma, to build a resource for rare histological subtypes of the lung and emphasize knowledge gaps in the management of these tumors. Our recommendations are based on a comprehensive review of case reports and series, clinical trials, and the "Indiana University Experience."
Collapse
Affiliation(s)
- Misty D Shields
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA
| | - Katherine G Minton
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA
| | - Mya Tran
- Department of Pharmacology and Toxicology, IUSOM
| | | | | | | | | | - Cynthia X Wei
- Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Julian A Marin-Acevedo
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA
| | - Rohan Maniar
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA
| | - Greg A Durm
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA
| | - Weston He
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA
| | - Nasser H Hanna
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine (IUSOM), Indianapolis, IN 46202, USA.
| |
Collapse
|
19
|
Rivas S, Sepúlveda RV, Tapia I, Estay C, Soto V, Blanco A, González E, Armisen R. MET Exon 14 Skipping and Novel Actionable Variants: Diagnostic and Therapeutic Implications in Latin American Non-Small-Cell Lung Cancer Patients. Int J Mol Sci 2024; 25:13715. [PMID: 39769478 PMCID: PMC11677537 DOI: 10.3390/ijms252413715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Targeted therapy indications for actionable variants in non-small-cell lung cancer (NSCLC) have primarily been studied in Caucasian populations, with limited data on Latin American patients. This study utilized a 52-genes next-generation sequencing (NGS) panel to analyze 1560 tumor biopsies from NSCLC patients in Chile, Brazil, and Peru. The RNA sequencing reads and DNA coverage were correlated to improve the detection of the actionable MET exon 14 skipping variant (METex14). The pathogenicity of MET variants of uncertain significance (VUSs) was assessed using bioinformatic methods, based on their predicted driver potential. The effects of the predicted drivers VUS T992I and H1094Y on c-MET signaling activation, proliferation, and migration were evaluated in HEK293T, BEAS-2B, and H1993 cell lines. Subsequently, c-Met inhibitors were tested in 2D and 3D cell cultures, and drug affinity was determined using 3D structure simulations. The prevalence of MET variants in the South American cohort was 8%, and RNA-based diagnosis detected 27% more cases of METex14 than DNA-based methods. Notably, 20% of METex14 cases with RNA reads below the detection threshold were confirmed using DNA analysis. The novel actionable T992I and H1094Y variants induced proliferation and migration through c-Met/Akt signaling. Both variants showed sensitivity to crizotinib and savolitinib, but the H1094Y variant exhibited reduced sensitivity to capmatinib. These findings highlight the importance of RNA-based METex14 diagnosis and reveal the drug sensitivity profiles of novel actionable MET variants from an understudied patient population.
Collapse
Affiliation(s)
- Solange Rivas
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| | - Romina V. Sepúlveda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Av. República 330, Santiago 8370146, Chile;
| | - Ignacio Tapia
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| | - Catalina Estay
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| | - Vicente Soto
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| | - Alejandro Blanco
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| | - Evelin González
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| | - Ricardo Armisen
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (I.T.); (C.E.); (V.S.); (A.B.); (E.G.)
| |
Collapse
|
20
|
Verkerk K, van der Wel TJWT, Zeverijn LJ, Geurts BS, Spiekman IAC, de Wit GF, Roepman P, Jansen AML, van der Noort V, Smit EF, Hoeben A, Hendriks LEL, van den Heuvel MM, Piet B, Herder GJM, Hashemi SMS, Gelderblom H, Verheul HMW, Voest EE, de Langen AJ. Safety, Efficacy, and Biomarker Analysis of Crizotinib in MET-Mutated Non-Small Cell Lung Cancer-Results from the Drug Rediscovery Protocol. Clin Cancer Res 2024; 30:5323-5332. [PMID: 39352721 DOI: 10.1158/1078-0432.ccr-24-1925] [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: 06/21/2024] [Revised: 08/26/2024] [Accepted: 09/26/2024] [Indexed: 12/12/2024]
Abstract
PURPOSE To provide patients with MET-mutated advanced non-small cell lung cancer (METmut aNSCLC) access to crizotinib, further substantiate evidence of its efficacy and safety in this setting, and find potential biomarkers for nonresponse. PATIENTS AND METHODS In the Drug Rediscovery Protocol (NCT0295234), patients with an actionable molecular profile are treated with off-label registered drugs. Both treated and untreated patients with aNSCLC harboring MET exon 14 skipping or other MET mutations received crizotinib 250 mg BID until disease progression or intolerable toxicity. Primary endpoints were clinical benefit [CB: RECIST v1.1 confirmed partial response, complete response (CR), or stable disease ≥16 weeks] and safety. Patients were enrolled using a Simon-like two-stage design, with eight patients in stage I and if ≥1/8 patients had CB, 24 patients in stage II. Whole-genome sequencing and RNA sequencing were performed on baseline biopsies. RESULTS Between September 2018 and October 2022, 30 patients started treatment, and 24 were response-evaluable after completing ≥1 full treatment cycle. Two patients (8.3%) achieved CR, 13 (54.2%) partial response, and two (8.3%) stable disease. The CB rate was 70.8% [95% confidence interval (CI), 48.9-87.4], and the objective response rate was 62.5% (95% CI, 40.6-81.2). After 21.2-month median follow-up, median duration of response, progression-free survival, and overall survival were 9.3 (95% CI, 6.5-not available), 10.2 (95% CI, 6.0-20.1), and 13.0 months (95% CI, 9.0-not available), respectively. Twenty-three treatment-related grade ≥ 3 adverse events occurred in 12/30 patients (40%), causing treatment discontinuation in three (10%). One patient (achieving CR) had a tyrosine kinase domain mutation (p.H1094Y), and all other patients had MET exon 14 skipping mutations. CONCLUSIONS Crizotinib is a valuable treatment option in METmut aNSCLC.
Collapse
Affiliation(s)
- Karlijn Verkerk
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | | | - Laurien J Zeverijn
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Birgit S Geurts
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Ilse A C Spiekman
- Oncode Institute, Utrecht, the Netherlands
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Gijs F de Wit
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Anne M L Jansen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Egbert F Smit
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ann Hoeben
- Division of Medical Oncology, Department of Internal Medicine, GROW School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonology, GROW - School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Michel M van den Heuvel
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Berber Piet
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gerarda J M Herder
- Department of Pulmonology, Meander Medical Center, Amersfoort, the Netherlands
| | - Sayed M S Hashemi
- Department of Pulmonary Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Emile E Voest
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Center for Personalized Cancer Treatment, the Netherlands
| | - Adrianus J de Langen
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
21
|
Mallareddy JR, Yang L, Lin WH, Feathers R, Ayers-Ringler J, Tolosa E, Kizhake AG, Kizhake S, Kubica SP, Boghean L, Alvarez S, Naldrett MJ, Singh S, Rana S, Zahid M, Schaefer-Klein J, Roden A, Kosari F, Anastasiadis PZ, Borad M, Natarajan A, Mansfield AS. Fluorescence based live cell imaging identifies exon 14 skipped hepatocyte growth factor receptor (MET) degraders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624922. [PMID: 39651287 PMCID: PMC11623520 DOI: 10.1101/2024.11.22.624922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Despite ongoing efforts to employ structure-based methods to discover targeted protein degraders (TPD), the prevailing strategy continues to be the synthesis of a focused set of heterobifunctional compounds and screen them for target protein degradation. Here we used a fluorescence based live cell imaging screen to identify degraders that target exon 14 skipped hepatocyte growth factor receptor (MET). MET is a known oncogenic driver. MET exon 14 skipping mutations (METex14Δ) are found in lung cancers and result in the loss of a degron that is required for E3-ligase recognition and subsequent ubiquitination, prolonging the half-life and oncogenicity of MET. Since proteolysis targeting chimeras (PROTACs) are heterobifunctional molecules that promote target degradation by the proteosome, we sought to restore degradation of MET lost with METex14Δ using a MET-targeting PROTAC. We generated a library of sixty PROTACs of which 37 used the MET inhibitor capmatinib as the protein of interest targeting ligand. We screened this PROTAC library for targeted degradation of METex14Δ-GFP using live cell imaging. We benchmarked out MET-targeting PROTACs to that of a previously reported MET-targeting PROTAC, SJF8240. Curve fitting live cell imaging data affords determination of time required to degrade 50% of the target protein (DT50), which was used in determining structure activity relationships. A promising candidate, 48-284, identified from the screen, exhibited classic PROTAC characteristics, was > 15-fold more potent than SJF8240, had fewer off targets compared to SJF8240, and degraded MET in multiple cell lines.
Collapse
|
22
|
Jiang Z, Gu Z, Yu X, Cheng T, Liu B. Research progress on the role of bypass activation mechanisms in resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Front Oncol 2024; 14:1447678. [PMID: 39582541 PMCID: PMC11581962 DOI: 10.3389/fonc.2024.1447678] [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: 06/12/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024] Open
Abstract
The clinical application of small molecule tyrosine kinase inhibitors (TKIs) has significantly improved the quality of life and prognosis of patients with non-small cell lung cancer (NSCLC) carrying driver genes. However, resistance to TKI treatment is inevitable. Bypass signal activation is one of the important reasons for TKI resistance. Although TKI drugs inhibit downstream signaling pathways of driver genes, key signaling pathways within tumor cells can still be persistently activated through bypass routes such as MET gene amplification, EGFR gene amplification, and AXL activation. This continuous activation maintains tumor cell growth and proliferation, leading to TKI resistance. The fundamental strategy to treat TKI resistance mediated by bypass activation involves simultaneously inhibiting the activated bypass signals and the original driver gene signaling pathways. Some clinical trials based on this combined treatment approach have yielded promising preliminary results, offering more treatment options for NSCLC patients with TKI resistance. Additionally, early identification of resistance mechanisms through liquid biopsy, personalized targeted therapy against these mechanisms, and preemptive targeting of drug-tolerant persistent cells may provide NSCLC patients with more sustained and effective treatment.
Collapse
Affiliation(s)
- Ziyang Jiang
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihan Gu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Yu
- Department of Emergency Medicine, West China Hospital, Sichuan University, West China School of Nursing, Sichuan University, Chengdu, China
- Institute of Disaster Medicine, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Chengdu, China
| | - Tao Cheng
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bofu Liu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Gallo S, Folco CB, Crepaldi T. The MET Oncogene: An Update on Targeting Strategies. Pharmaceuticals (Basel) 2024; 17:1473. [PMID: 39598385 PMCID: PMC11597589 DOI: 10.3390/ph17111473] [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: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The MET receptor, commonly known as HGF (hepatocyte growth factor) receptor, is a focus of extensive scientific research. MET has been linked to embryonic development, tissue regeneration following injury, tumorigenesis, and cancer metastasis. These functions underscore its involvement in numerous cellular processes, including stemness, proliferation, motility, cell dissociation, and survival. However, the enigmatic nature of MET becomes apparent in the context of cancer. When MET remains persistently activated, since its gene undergoes genetic alterations, it initiates a complex signaling cascade setting in motion an aggressive and metastatic program that is characteristic of malignant cells and is known as "invasive growth". The expanding knowledge of MET signaling has opened up numerous opportunities for therapeutic interventions, particularly in the realm of oncology. Targeting MET presents a promising strategy for developing novel anti-cancer treatments. In this review, we provide an updated overview of drugs designed to modulate MET signaling, highlighting MET kinase inhibitors, degraders, anti-MET/HGF monoclonal antibodies, and MET-targeted antibody-drug conjugates. Through this review, we aim to contribute to the ongoing advancement of therapeutic strategies targeting MET signaling.
Collapse
Affiliation(s)
- Simona Gallo
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Consolata Beatrice Folco
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| |
Collapse
|
24
|
Li R, Liu X, Xu Y, Zhao J, Zhong W, Gao X, Chen M, Wang M. Remarkable pathological response to neoadjuvant tepotinib in lung adenocarcinoma with MET exon 14 skipping mutation: A case report. Thorac Cancer 2024; 15:2339-2343. [PMID: 39343987 PMCID: PMC11554545 DOI: 10.1111/1759-7714.15459] [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: 06/20/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Mesenchymal-epithelial transition (MET) exon 14 (METex14) skipping mutation is a rare (3%-4%) driver mutation in non-small cell lung cancer (NSCLC). Tepotinib, a selective MET inhibitor, has shown promise in treating METex14 skipping-mutated NSCLC. However, its feasibility for perioperative application remains unclear. This report describes a 60-year-old man with stage IIIA (cT2N2M0) lung adenocarcinoma harboring a METex14 skipping mutation. After initial treatment with savolitinib was discontinued due to grade 4 transaminitis, the patient was switched to tepotinib, resulting in significant tumor regression. Six months later, further shrinkage was observed, and surgery revealed remarkable pathological response with no residual tumor in lymph nodes (ypT2N0M0, IB). Postoperative tepotinib continued, with no relapse at 6-month follow-up. This case highlights the potential of tepotinib as neoadjuvant therapy for resectable METex14 skipping-mutated NSCLC, warranting further clinical trials.
Collapse
Affiliation(s)
- Rongzhen Li
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yan Xu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jing Zhao
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wei Zhong
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoxing Gao
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Minjiang Chen
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mengzhao Wang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
25
|
Wang K, Hsu R. Anti-MET Antibody Therapies in Non-Small-Cell Lung Cancer: Current Progress and Future Directions. Antibodies (Basel) 2024; 13:88. [PMID: 39449330 PMCID: PMC11503282 DOI: 10.3390/antib13040088] [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: 09/01/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Non-small-cell lung cancer (NSCLC) remains a leading cause of cancer mortality globally, though advances in targeted therapies have improved treatment outcomes. The mesenchymal-epithelial transition (MET) gene plays a significant role in NSCLC, often through protein overexpression, exon 14 skipping mutations, and gene amplification, many of which arise as resistance mechanisms to other oncogenic drivers like epidermal growth factor receptor (EGFR) mutations. This review examines the development and clinical efficacy of anti-MET antibody therapies. Methods: A comprehensive literature search was conducted using major medical databases looking at key relevant studies on anti-MET antibody studies. Both authors reviewed the literature, assessed study quality, and interpreted the results from each study. Results: Amivantamab, a bispecific EGFR/MET antibody was approved to treat EGFR exon 20 insertion and now has recently been extended to target classical EGFR mutations with progression on osimertinib. Other important anti-MET targeted therapies in development include antibody drug conjugates such as telisotuzumab vedotin, REGN5093-M114, and AZD9592 and emibetuzumab, which is a humanized immunoglobulin G4 monoclonal bivalent MET antibody. Conclusions: MET plays a significant role in NSCLC and amivantamab along with other anti-MET targeted therapies play a role in directly targeting MET and addressing acquired resistance to oncogenic drivers. Future research should focus on developing novel MET antibody drugs and exploring new therapeutic combinations to enhance treatment efficacy and overcome resistance in NSCLC. Refining biomarker-driven approaches to ensure precise patient selection is also critical to optimizing treatment outcomes.
Collapse
Affiliation(s)
- Kinsley Wang
- Department of Medicine, University of Arizona College of Medicine—Phoenix, Phoenix, AZ 85004, USA;
| | - Robert Hsu
- Department of Medicine, Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| |
Collapse
|
26
|
Ye L, Wang W, Li H, Ji Y, Le X, Xu X. Targeting the MET gene: unveiling therapeutic opportunities in immunotherapy within the tumor immune microenvironment of non-small cell lung cancer. Ther Adv Med Oncol 2024; 16:17588359241290733. [PMID: 39483139 PMCID: PMC11526239 DOI: 10.1177/17588359241290733] [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: 07/13/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) represents the most prevalent histological subtype of lung cancer. Within this disease, the MET gene emerges as a critical therapeutic target, exhibiting various forms of dysregulation. Although MET tyrosine kinase inhibitors, HGF/c-MET targeting antibodies, and antibody-drug conjugates constitute the primary treatment modalities for patients with MET-altered NSCLC, numerous questions remain regarding their optimal application. The advent of immunotherapy holds promise for enhancing therapeutic outcomes in patients with MET-altered NSCLC. MET mutations can reshape the tumor immune microenvironment of NSCLC by reducing tumor immunogenicity, inducing exhaustion in immune-activated cells, and promoting immune evasion, which are crucial for modulating treatment responses. Furthermore, we emphasize the promising synergy of immunotherapy with emerging treatments and the challenges and opportunities in refining these approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Lisha Ye
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Wenjing Wang
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Huihui Li
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yongling Ji
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 Banshan East Road, Hangzhou, Zhejiang 310022, China
| | - Xiuning Le
- Division of Internal Medicine, Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4000, USA
| | - Xiaoling Xu
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Zhenmin Road, Shanghai 200433, China
| |
Collapse
|
27
|
Wolf J, Hochmair M, Han JY, Reguart N, Souquet PJ, Smit EF, Orlov SV, Vansteenkiste J, Nishio M, de Jonge M, Akerley W, Garon EB, Groen HJM, Tan DSW, Seto T, Frampton GM, Robeva A, Carbini M, Le Mouhaer S, Yovine A, Boran A, Bossen C, Yang Y, Ji L, Fairchild L, Heist RS. Capmatinib in MET exon 14-mutated non-small-cell lung cancer: final results from the open-label, phase 2 GEOMETRY mono-1 trial. Lancet Oncol 2024; 25:1357-1370. [PMID: 39362249 DOI: 10.1016/s1470-2045(24)00441-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Capmatinib has previously shown activity in treatment-naive and previously treated patients with non-small-cell lung cancer (NSCLC) and a MET exon 14-skipping mutation (METex14). Here, we report the final outcomes from the phase 2 GEOMETRY mono-1 study with an aim to provide further evidence for the activity of capmatinib. METHODS In this non-randomised, multi-cohort, open-label, phase 2 trial conducted in 152 centres and hospitals in 25 countries, with patients treated in 95 centres in 20 countries, eligible patients (aged ≥18 years) with MET-dysregulated, EGFR wild-type, and ALK rearrangement-negative advanced NSCLC (stage IIIB/IV) and an Eastern Cooperative Oncology Group performance status of 0 or 1 were assigned to cohorts (1a, 1b, 2, 3, 4, 5a, 5b, 6 and 7) based on their MET status (METex14 or MET amplification) and previous therapy lines. Patients received capmatinib (400 mg orally twice daily) in 21-day treatment cycles. The primary endpoint was overall response rate by blinded independent central review per Response Evaluation Criteria in Solid Tumours version 1.1 and was performed on the full analysis set (all patients who received at least one dose of capmatinib). Previous reports of this study had published interim or primary data for cohorts 1-7. Here, we report the final clinical outcomes from all METex14 cohorts (4, 5b, 6, and 7) and safety from all study cohorts (1-7). The trial is registered with ClinicalTrials.gov, NCT02414139, and has been completed. FINDINGS Of 373 treated patients enrolled from June 11, 2015, to March 12, 2020, 160 (97 [61%] female) patients had METex14 NSCLC and were enrolled in four cohorts: 60 treatment-naive (cohorts 5b and 7) and 100 previously treated (cohorts 4 and 6). The overall median study follow-up was 46·4 months (IQR 41·8-65·4) for the treatment-naïve patients and 66·9 months (56·7-73·9) for previously treated patients, respectively. Overall responses were recorded in 41 (68%; 95% CI 55·0-79·7) of 60 treatment-naive patients and 44 (44%; 95% CI 34·1-54·3) of 100 previously treated patients. In all 373 treated patients, the most common treatment-related adverse events were peripheral oedema (n=174; 47%), nausea (n=130; 35%), increased blood creatinine (n=78; 21%), and vomiting (n=74; 20%). Grade 3-4 serious adverse events occurred in 164 (44%) patients, dyspnoea being the most common (18 patients [5%]). Treatment-related deaths occurred in four (1%) patients (one each of cardiac arrest, hepatitis, organising pneumonia, and pneumonitis). No new safety signals were reported. INTERPRETATION These long-term results support METex14 as a targetable oncogenic driver in NSCLC and add to the evidence supporting capmatinib as a targeted treatment option for treatment-naive and previously treated patients with METex14 NSCLC. FUNDING Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Jürgen Wolf
- Department of Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany.
| | - Maximilian Hochmair
- Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Department of Respiratory and Critical Care Medicine, Krankenhaus Nord, Klinik Floridsdorf, Vienna, Austria
| | | | - Noemi Reguart
- Medical Oncology Department, Hospital Clínic i Provincial de Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumours, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pierre-Jean Souquet
- Department of Thoracic Oncology, CH Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Egbert F Smit
- Department of Pulmonary Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Sergey V Orlov
- Department of Thoracic Oncology, St Petersburg State Medical University, St Petersburg, Russia
| | | | - Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Maja de Jonge
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Wallace Akerley
- Huntsman Cancer Institute, Salt Lake City, UT, USA; Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Edward B Garon
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Harry J M Groen
- Department of Pulmonary Diseases, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Daniel S W Tan
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore
| | | | | | - Anna Robeva
- Oncology Global Drug Development, Novartis Pharmaceuticals, East Hanover, NJ, USA
| | - Mariana Carbini
- Oncology DU Global Drug Development, Novartis Pharma, Basel, Switzerland
| | - Sylvie Le Mouhaer
- Global Drug Development, Novartis Pharma S.A.S, Rueil Malmaison, France
| | - Alejandro Yovine
- Oncology DU Global Drug Development, Novartis Pharma, Basel, Switzerland
| | - Aislyn Boran
- Global Drug Development, Novartis Services, East Hanover, New Jersey, USA
| | - Claudia Bossen
- Oncology DU Global Drug Development, Novartis Pharma, Basel, Switzerland
| | - Yiqun Yang
- Oncology Global Drug Development, Novartis Pharmaceuticals, East Hanover, NJ, USA
| | - Lexiang Ji
- Oncology Data Science, Novartis BioMedical Research, Cambridge, MA, USA
| | - Lauren Fairchild
- Oncology Data Science, Novartis BioMedical Research, Cambridge, MA, USA
| | - Rebecca S Heist
- Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
28
|
Wang Y, Wei J, Xu M, Xiang J, Shao K, Hao Y, Song Z. Efficacy and safety analysis of immunotherapy in non-small cell lung cancer patients with MET alterations. Clin Transl Oncol 2024; 26:2503-2512. [PMID: 38627317 DOI: 10.1007/s12094-024-03455-y] [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: 01/27/2024] [Accepted: 03/06/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Mesenchymal epithelial transition factor (MET) is a rare oncologic driver gene, and information on immunotherapy for non-small cell lung cancer (NSCLC) patients with this driver gene is limited. Here we evaluate the efficacy and safety of immune checkpoint inhibitors (ICI) under different therapeutic regimen for NSCLC patients with MET alterations. METHODS From June 2019 to December 2023, we assessed the efficacy and toxicity of ICIs in 42 NSCLC patients with MET alterations. Survival curves were plotted using the Kaplan-Meier method and the Cox proportional hazards model applied for univariate and multivariate analyses. We assessed the size of target lesion according to RECIST v1.1, and objective response rate (ORR) was defined as the sum of complete response (CR) and partial response (PR), disease control rate (DCR) as the sum of CR, PR, and disease stable. RESULTS A total of 42 NSCLC patients with MET alterations were included in this retrospective study, 10 was MET 14 skipping mutation and 32 was MET amplification. The ORR for ICI treatment was 30.95% and the DCR was 71.43%. Median progression-free survival (mPFS) and median overall survival (OS) were 4.40 and 13.97 months, respectively. There exists statistical differences between the mPFS of ICI monotherapy and combine ICI therapy (2.8 vs 7.8 months, p = 0.022). The incidence of drug-related adverse reactions was 47.62%, mainly bone marrow suppression (14.28%), immune-related pneumonia (7.14%), and liver function impairment (7.14%), and six patients (14.28%) experiencing grade 3 or above adverse events. CONCLUSION NSCLC patients with MET alterations can benefit from immunotherapy, especially the patients treated by combined ICI therapy. However, special attention should be paid to the occurrence of grade 3/4 adverse reactions while using the combined ICI therapy.
Collapse
Affiliation(s)
- Yanhua Wang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310000, China
- Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Jingwen Wei
- Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Manyi Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310000, China
- Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Jing Xiang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310000, China
- Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Keda Shao
- Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Yue Hao
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310000, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China.
- Department of Clinical Trial, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
| | - Zhengbo Song
- Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China.
- Department of Clinical Trial, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
29
|
Chen SY, Wang CT, Huang TH, Tsai JL, Wang HT, Yen YT, Tseng YL, Wu CL, Chang JM, Shiau AL. Advancing Lung Cancer Treatment with Combined c-Met Promoter-Driven Oncolytic Adenovirus and Rapamycin. Cells 2024; 13:1597. [PMID: 39329778 PMCID: PMC11430802 DOI: 10.3390/cells13181597] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Lung cancer remains a formidable health challenge due to its high mortality and morbidity rates. Non-small cell lung cancer (NSCLC) constitutes approximately 85% of all lung cancer cases, with small cell lung cancer (SCLC) accounting for the remainder. Both NSCLC and SCLC cells express receptor tyrosine kinases, which may be overexpressed or mutated in lung cancer, leading to increased activation. The c-Met receptor tyrosine kinase, crucial for cell transformation and tumor growth, invasion, and metastasis, became the focus of our study. We used an E1B55KD-deleted, replication-selective oncolytic adenovirus (Ad.What), driven by the c-Met promoter, targeting lung cancer cells with c-Met overexpression, thus sparing normal cells. Previous studies have shown the enhanced antitumor efficacy of oncolytic adenoviruses when combined with chemotherapeutic agents. We explored combining rapamycin, a selective mTOR inhibitor with promising clinical trial outcomes for various cancers, with Ad.What. This combination increased infectivity by augmenting the expression of coxsackievirus and adenovirus receptors and αV integrin on cancer cells and induced autophagy. Our findings suggest that combining a c-Met promoter-driven oncolytic adenovirus with rapamycin could be an effective lung cancer treatment strategy, offering a targeted approach to exploit lung cancer cells' vulnerabilities, potentially marking a significant advancement in managing this deadly disease.
Collapse
Affiliation(s)
- Shih-Yao Chen
- Department of Nursing, College of Nursing, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan;
| | - Chung-Teng Wang
- Tong Yuan Diabetes Center, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (C.-T.W.); (C.-L.W.)
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tang-Hsiu Huang
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Jeng-Liang Tsai
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hao-Tien Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Ting Yen
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-T.Y.); (Y.-L.T.)
| | - Yau-Lin Tseng
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-T.Y.); (Y.-L.T.)
| | - Chao-Liang Wu
- Tong Yuan Diabetes Center, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (C.-T.W.); (C.-L.W.)
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan
| | - Jia-Ming Chang
- Thoracic Division, Department of Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan
- Institute of Molecular Biology, National Chung Cheng University, Chiayi 62102, Taiwan
| | - Ai-Li Shiau
- Tong Yuan Diabetes Center, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (C.-T.W.); (C.-L.W.)
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
30
|
Kawczak P, Feszak I, Bączek T. Epinephrine, Pregabalin, and Crizotinib as Three Medicines with Polish Implications over Three Last Centuries and in View of Three Different Drug Discovery Approaches. Biomedicines 2024; 12:2021. [PMID: 39335535 PMCID: PMC11428485 DOI: 10.3390/biomedicines12092021] [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: 07/21/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The discovery of epinephrine (adrenaline) and its subsequent implications in medicine owes significant contributions to Cybulski across different centuries, who, in 1894, was pivotal in identifying the adrenal medulla's role in blood pressure regulation and naming the active substance "nadnerczyna", known today as adrenaline. His work demonstrated the adrenal glands' critical function in the body's regulatory mechanisms beyond the nervous system. Cybulski's groundbreaking research laid foundational knowledge for future endocrinological studies and pharmaceutical advancements. In the late 20th century, Andruszkiewicz collaborated with Silverman at Northwestern University to develop pregabalin, the active ingredient in Lyrica. Their innovative synthesis of gamma-aminobutyric acid derivatives led to a significant advancement in treating epilepsy, neuropathic pain, and fibromyalgia. Andruszkiewicz's expertise in organic chemistry and enzymology was crucial in this collaborative effort, resulting in the successful development and commercialization of Lyrica. Additionally, Mroczkowski's leadership at Pfizer contributed to the development of crizotinib, a notable anaplastic lymphoma kinase and proto-oncogene 1 tyrosine-protein kinase inhibitor used to treat specific types of non-small cell lung cancer. Her work exemplifies the continuing influence of Polish researchers in pioneering drug discovery and advancing therapeutic treatments over the past three centuries. These contributions highlight Poland's significant role in global pharmaceutical innovations and medical research.
Collapse
Affiliation(s)
- Piotr Kawczak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland;
| | - Igor Feszak
- Institute of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland;
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland;
- Department of Nursing and Medical Rescue, Institute of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland
| |
Collapse
|
31
|
Minne RL, Luo NY, Traynor AM, Huang M, DeTullio L, Godden J, Stoppler M, Kimple RJ, Baschnagel AM. Genomic and Immune Landscape Comparison of MET Exon 14 Skipping and MET-Amplified Non-small Cell Lung Cancer. Clin Lung Cancer 2024; 25:567-576.e1. [PMID: 38852006 DOI: 10.1016/j.cllc.2024.05.001] [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: 01/22/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Mutation or amplification of the mesenchymal-epithelial transition (MET) tyrosine kinase receptor causes dysregulation of receptor function and stimulates tumor growth in non-small cell lung cancer (NSCLC) with the most common mutation being MET exon 14 (METex14). We sought to compare the genomic and immune landscape of MET-altered NSCLC with MET wild-type NSCLC. METHODS 18,047 NSCLC tumors were sequenced with Tempus xT assay. Tumors were categorized based on MET exon 14 (METex14) mutations; low MET amplification defined as a copy number gain (CNG) 6-9, high MET amplification defined as CNG ≥ 10, and MET other type mutations. Immuno-oncology (IO) biomarkers and the frequency of other somatic gene alterations were compared across MET-altered and MET wild-type groups. RESULTS 276 (1.53%) METex14, 138 (0.76%) high METamp, 63 (0.35%) low METamp, 27 (0.15%) MET other, and 17,543 (97%) MET wild-type were identified. Patients with any MET mutation including METex14 were older, while patients with METex14 were more frequently female and nonsmokers. MET gene expression was highest in METamp tumors. PD-L1 positivity rates were higher in MET-altered groups than MET wild-type. METex14 exhibited the lowest tumor mutational burden (TMB) and lowest neoantigen tumor burden (NTB). METamp exhibited the lowest proportion of CD4 T cells and the highest proportion of NK cells. There were significant differences in co-alterations between METamp and METex14. CONCLUSIONS METex14 tumors exhibited differences in IO biomarkers and the somatic landscape compared to non-METex14 NSCLC tumors. Variations in immune profiles can affect immunotherapy selection in MET-altered NSCLC and require further exploration.
Collapse
Affiliation(s)
- Rachel L Minne
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Natalie Y Luo
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Anne M Traynor
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | | | | | | | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Andrew M Baschnagel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
32
|
Bontoux C, Hofman V, Chamorey E, Schiappa R, Lassalle S, Long-Mira E, Zahaf K, Lalvée S, Fayada J, Bonnetaud C, Goffinet S, Ilié M, Hofman P. Reproducibility of c-Met Immunohistochemical Scoring (Clone SP44) for Non-Small Cell Lung Cancer Using Conventional Light Microscopy and Whole Slide Imaging. Am J Surg Pathol 2024; 48:1072-1081. [PMID: 38980727 DOI: 10.1097/pas.0000000000002274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Emerging therapies for non-small cell lung cancer targeting c-Met overexpression have recently demonstrated promising results. However, the evaluation of c-Met expression can be challenging. We aimed to study the inter and intraobserver reproducibility of c-Met expression evaluation. One hundred ten cases with non-small cell lung cancer (40 biopsies and 70 surgical specimens) were retrospectively selected in a single laboratory (LPCE) and evaluated for c-Met expression. Six pathologists (4 seniors and 2 juniors) evaluated the H-score and made a 3-tier classification of c-Met expression for all cases, using conventional light microscopy (CLM) and whole slide imaging (WSI). The interobserver reproducibility with CLM gave global Cohen Kappa coefficients (ƙ) ranging from 0.581 (95% CI: 0.364-0.771) to 0.763 (95% CI: 0.58-0.92) using the c-Met 3-tier classification and H-score, respectively. ƙ was higher for senior pathologists and biopsy samples. The interobserver reproducibility with WSI gave a global ƙ ranging from 0.543 (95% CI: 0.33-0.724) to 0.905 (95% CI: 0.618-1) using the c-Met H-score and 2-tier classification (≥25% 3+), respectively. ƙ for intraobserver reproducibility between CLM and WSI ranged from 0.713 to 0.898 for the c-Met H-score and from 0.600 to 0.779 for the c-Met 3-tier classification. We demonstrated a moderate to excellent interobserver agreement for c-Met expression with a substantial to excellent intraobserver agreement between CLM and WSI, thereby supporting the development of digital pathology. However, some factors (scoring method, type of tissue samples, and expertise level) affect reproducibility. Our findings highlight the importance of establishing a consensus definition and providing further training, particularly for inexperienced pathologists, for c-Met immunohistochemistry assessment in clinical practice.
Collapse
Affiliation(s)
- Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- Team 4, Institute of Research on Cancer and Aging of Nice Inserm U1081, CNRS UMR7284, Côte d'Azur University
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- Team 4, Institute of Research on Cancer and Aging of Nice Inserm U1081, CNRS UMR7284, Côte d'Azur University
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Emmanuel Chamorey
- Department of Statistics, Antoine Lacassagne Cancer Center, Nice, France
| | - Renaud Schiappa
- Department of Statistics, Antoine Lacassagne Cancer Center, Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- Team 4, Institute of Research on Cancer and Aging of Nice Inserm U1081, CNRS UMR7284, Côte d'Azur University
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- Team 4, Institute of Research on Cancer and Aging of Nice Inserm U1081, CNRS UMR7284, Côte d'Azur University
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Julien Fayada
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Christelle Bonnetaud
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | | | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- Team 4, Institute of Research on Cancer and Aging of Nice Inserm U1081, CNRS UMR7284, Côte d'Azur University
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology
- Hospital-Integrated Biobank
- Team 4, Institute of Research on Cancer and Aging of Nice Inserm U1081, CNRS UMR7284, Côte d'Azur University
- FHU OncoAge, Côte d'Azur University
- University Hospital Institute RespirERA, Côte d'Azur University, Pasteur Hospital, CHU of Nice
| |
Collapse
|
33
|
Zhu J, Chen J, Liu W, Zhang J, Gu Y. Mutation of MET D1228N as an Acquired Potential Mechanism of Crizotinib Resistance in NSCLC with MET Y1003H Mutation. LUNG CANCER (AUCKLAND, N.Z.) 2024; 15:123-128. [PMID: 39221108 PMCID: PMC11365520 DOI: 10.2147/lctt.s467584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Mesenchymal-epithelial transition (MET) gene has been identified as a promising target for treatments. However, different sites of the MET mutation show different effects to MET inhibition. Here, we reported a non-small cell lung cancer (NSCLC) patient harboring MET Y1003H mutation who achieved a durable partial response to crizotinib with a PFS of 22.4 months. Furthermore, we report for the first time the identification of MET D1228N as a possible mechanism of acquired resistance to crizotinib in a patient with MET Y1003H mutation during disease progression.
Collapse
Affiliation(s)
- Jinlian Zhu
- Department of Oncology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu Province, People’s Republic of China
| | - Jie Chen
- Department of Oncology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu Province, People’s Republic of China
| | - Wei Liu
- Department of Oncology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu Province, People’s Republic of China
| | - Junling Zhang
- Medical Department, 3D Medicines Inc, Shanghai, People’s Republic of China
| | - Yulan Gu
- Department of Oncology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu Province, People’s Republic of China
| |
Collapse
|
34
|
Yao S, Liu X, Feng Y, Li Y, Xiao X, Han Y, Xia S. Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment. Int J Mol Sci 2024; 25:9101. [PMID: 39201787 PMCID: PMC11354629 DOI: 10.3390/ijms25169101] [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: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is characterized by several molecular alterations that contribute to its development and progression. These alterations include the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), human epidermal growth factor receptor 2 (HER2), and mesenchymal-epithelial transition factor (c-MET). Among these, the hepatocyte growth factor (HGF)/c-MET signaling pathway plays a crucial role in NSCLC. In spite of this, the involvement of the HGF/c-MET signaling axis in remodeling the tumor microenvironment (TME) remains relatively unexplored. This review explores the biological functions of the HGF/c-MET signaling pathway in both normal and cancerous cells, examining its multifaceted roles in the NSCLC tumor microenvironment, including tumor cell proliferation, migration and invasion, angiogenesis, and immune evasion. Furthermore, we summarize the current progress and clinical applications of MET-targeted therapies in NSCLC and discuss future research directions, such as the development of novel MET inhibitors and the potential of combination immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (S.Y.); (X.L.); (Y.F.); (Y.L.); (X.X.); (Y.H.)
| |
Collapse
|
35
|
Nosaki K, Yoh K, Toyozawa R, Horinouchi H, Morise M, Ohashi K, Murakami H, Satouchi M, Sakakibara-Konishi J, Yano S, Okumura F, Matsumoto S, Shimokawa M, Seto T, Goto K. Phase 2 trial of crizotinib in Japanese patients with advanced NSCLC harboring a MET gene alteration: a Co-MET study. Int J Clin Oncol 2024; 29:1142-1151. [PMID: 38758397 DOI: 10.1007/s10147-024-02543-x] [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: 01/15/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND MET exon 14 skipping mutations occur in 3-4% and MET high amplifications occur in < 1% of patients with non-small-cell lung cancer (NSCLC). Crizotinib, a selective ATP-competitive small-molecule inhibitor of c-Met, ALK, and ROS1 tyrosine kinases, has shown activity in cancer models with various types of MET activation. METHODS The Co-MET study is a single-arm phase 2 trial to assess the safety and efficacy of crizotinib in MET inhibitor-naïve patients with advanced NSCLC harboring MET exon 14 skipping mutation (cohort 1) or high MET gene copy number of ≥ 7 (cohort 2). The primary endpoint was the objective response rate (ORR) per RECIST v1.1 by independent radiology review in cohort 1. The key secondary endpoints were the duration of response (DoR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS A total of 28 patients (23 in cohort 1 and 5 in cohort 2) were enrolled between March 2018 and February 2020. The primary endpoint was met as the ORR (90% confidence interval: CI) in cohort 1 was 38.1% (20.6-58.3). Median DoR, PFS, and OS (95% CI) were 7.6 (1.9-NE), 5.7 (2.1-11.3), 9.1 (4.0-19.9) months, respectively, in cohort 1. ORR in cohort 2 was 40.0% (18.9-92.4). The safety signals were generally consistent with the known safety profile of crizotinib. CONCLUSIONS Crizotinib showed a clinical activity similar to that of tepotinib and capmatinib in patients with NSCLC harboring MET exon 14 skipping mutations. CLINICAL TRIAL INFORMATION UMIN000031623.
Collapse
Affiliation(s)
- Kaname Nosaki
- Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Ryo Toyozawa
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kadoaki Ohashi
- Department of Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Haruyasu Murakami
- Division of Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Miyako Satouchi
- Department of Thoracic Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Seiji Yano
- Department of Respiratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Fumihiko Okumura
- Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Seto
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| |
Collapse
|
36
|
Chen S, Hu T, Zhao J, Zhu Q, Wang J, Huang Z, Xiang C, Zhao R, Zhu C, Lu S, Han Y. Novel molecular subtypes of METex14 non-small cell lung cancer with distinct biological and clinical significance. NPJ Precis Oncol 2024; 8:159. [PMID: 39060379 PMCID: PMC11282101 DOI: 10.1038/s41698-024-00642-6] [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: 12/29/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Not all MET exon 14 skipping (METex14) NSCLC patients benefited from MET inhibitors. We hypothesized an inter-tumoral heterogeneity in METex14 NSCLC. Investigations at genomic and transcriptomic level were conducted in METex14 NSCLC samples from stage I-III and recurrent/metastatic patients as discovery and validation cohort. Four molecular subtypes were discovered. MET-Driven subtype, with the worst prognosis, displayed MET overexpression, enrichment of MET-related pathways, and higher infiltration of fibroblast and regulatory T cells. Immune-Activated subtype having the most idea long-term survival, had higher tertiary lymphoid structures, spatial co-option of PD-L1+ cancer cells, and GZMK+ CD8+ T cell. FGFR- and Bypass-Activated subtypes displayed FGFR2 overexpression and enrichments of multiple oncogenic pathways respectively. In the validation cohort, patients with MET-Driven subtype had better response to MET inhibitors than those with MET overexpression. Thus, molecular subtypes of METex14 NSCLC with distinct biological and clinical significance may indicate more precise therapeutic strategies for METex14 NSCLC patients.
Collapse
Affiliation(s)
- Shengnan Chen
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tao Hu
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Jikai Zhao
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qian Zhu
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jin Wang
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Zhan Huang
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Chan Xiang
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ruiying Zhao
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Changbin Zhu
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China.
| | - Shun Lu
- Department of Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
37
|
Zhang H, Zhang Y, Zhu Y, Dong T, Liu Z. Understanding the treatment response and resistance to targeted therapies in non-small cell lung cancer: clinical insights and perspectives. Front Oncol 2024; 14:1387345. [PMID: 39055566 PMCID: PMC11269125 DOI: 10.3389/fonc.2024.1387345] [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: 02/17/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Lung cancer remains the leading cause of mortality worldwide. Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer with a generally poor prognosis. In recent years, advances in targeted therapy and sequencing technology have brought significant improvement in the therapeutic outcomes of patients with advanced NSCLC. Targeted inhibitors directed against specific mutated or rearranged oncogenes, such as epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), and receptor tyrosine kinase ROS proto-oncogene 1(ROS1) among others, exhibit promising anti-tumor activity. Unfortunately, some patients develop acquired resistance and disease progression soon after initial remission. Despite the continuous development of new drugs and strategies to overcome drug resistance, it is still a major challenge in the treatment of NSCLC. The landscape of targeted therapy for NSCLC is evolving rapidly in response to the pace of scientific research. This study aimed to provide a comprehensive review of tumor target antigens and agents related to targeted therapy in NSCLC.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Yingying Zhang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Yingying Zhu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Dong
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Zheng Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Sposito M, Belluomini L, Nocini R, Insolda J, Scaglione IM, Menis J, Simbolo M, Lugini A, Buzzacchino F, Verderame F, Spinnato F, Aprile G, Calvetti L, Occhipinti M, Marinelli D, Veccia A, Lombardo F, Soto Parra HJ, Ferraù F, Savastano C, Porta C, Pradelli L, Sicari E, Castellani S, Malapelle U, Novello S, Bria E, Pilotto S, Milella M. Tissue- and liquid-biopsy based NGS profiling in advanced non-small-cell lung cancer in a real-world setting: the IMMINENT study. Front Oncol 2024; 14:1436588. [PMID: 39045557 PMCID: PMC11263796 DOI: 10.3389/fonc.2024.1436588] [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: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction To date, for all non-small cell lung cancer (NSCLC) cases, it is recommended to test for driver alterations to identify actionable therapeutic targets. In this light, comprehensive genomic profiling (CGP) with next generation sequencing (NGS) has progressively gained increasing importance in clinical practice. Here, with the aim of assessing the distribution and the real-world frequency of gene alterations and their correlation with patient characteristics, we present the outcomes obtained using FoundationOne (F1CDx) and FoundationLiquid CDx (F1L/F1LCDx) NGS-based profiling in a nationwide initiative for advanced NSCLC patients. Methods F1CDx (324 genes) was used for tissue samples, and F1L (70 genes) or F1LCDx (324 genes) for liquid biopsy, aiming to explore the real-world occurrence of molecular alterations in aNSCLC and their relationship with patients' characteristics. Results Overall, 232 advanced NSCLC patients from 11 Institutions were gathered [median age 63 years; never/former or current smokers 29.3/65.9%; adenocarcinoma/squamous 79.3/12.5%; F1CDx/F1L+F1LCDx 59.5/40.5%]. Alterations were found in 170 different genes. Median number of mutated genes per sample was 4 (IQR 3-6) and 2 (IQR 1-3) in the F1CDx and F1L/F1LCDx cohorts, respectively. TP53 (58%), KRAS (22%), CDKN2A/B (19%), and STK11 (17%) alterations were the most frequently detected. Actionability rates (tier I and II) were comparable: 36.2% F1CDx vs. 34% ctDNA NGS assays (29.5% and 40.9% F1L and F1LCDx, respectively). Alterations in KEAP1 were significantly associated with STK11 and KRAS, so as TP53 with RB1. Median tumor mutational burden was 6 (IQR 3-10) and was significantly higher in smokers. Median OS from metastatic diagnosis was 23 months (IQR 18.5-19.5) and significantly lower in patients harboring ≥3 gene mutations. Conditional three-year survival probabilities increased over time for patients profiled at initial diagnosis and exceeded those of individuals tested later in their clinical history after 12 months. Conclusion This study confirms that NGS-based molecular profiling of aNSCLC on tissue or blood samples offers valuable predictive and prognostic insights.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Lorenzo Belluomini
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Riccardo Nocini
- Otolaryngology-Head and Neck Surgery Department, University of Verona Hospital Trust, Verona, Italy
| | - Jessica Insolda
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Ilaria Mariangela Scaglione
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Jessica Menis
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Antonio Lugini
- Medical Oncology Unit, Azienda Ospedaliera (AO) San Giovanni Addolorata Hospital, Rome, Italy
| | | | - Francesco Verderame
- Section of Oncology, Azienda Ospedaliera (AO) Ospedali Riuniti “Villa Sofia- V. Cervello”, Palermo, Italy
| | - Francesca Spinnato
- Section of Oncology, Azienda Ospedaliera (AO) Ospedali Riuniti “Villa Sofia- V. Cervello”, Palermo, Italy
| | - Giuseppe Aprile
- Department of Clinical Oncology, San Bortolo General Hospital, Azienda ULSS8 Berica, Vicenza, Italy
| | - Lorenzo Calvetti
- Department of Clinical Oncology, San Bortolo General Hospital, Azienda ULSS8 Berica, Vicenza, Italy
| | - Mario Occhipinti
- Department of Experimental Medicine, Sapienza University, Rome, Italy
- Medical Oncology Department, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Dei Tumori, Milan, Italy
| | - Daniele Marinelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
- Division of Medical Oncology B, Policlinico Umberto I, Rome, Italy
| | - Antonello Veccia
- Medical Oncology Department, Santa Chiara Hospital, Trento, Italy
| | | | - Hector José Soto Parra
- Medical Oncology, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-S. Marco”, Catania, Italy
| | - Francesco Ferraù
- Department of Medical Oncology, Unità Operativa Complessa (UOC) Oncologia, Taormina, Italy
| | | | - Camilla Porta
- AdRes Health Economics and Outcome Research, Turin, Italy
| | | | | | | | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Silvia Novello
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Sara Pilotto
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Michele Milella
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| |
Collapse
|
39
|
Lieber A, Makai A, Orosz Z, Kardos T, Isaac SJ, Tornyi I, Bittner N. The role of immunotherapy in early-stage and metastatic NSCLC. Pathol Oncol Res 2024; 30:1611713. [PMID: 39027681 PMCID: PMC11254634 DOI: 10.3389/pore.2024.1611713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024]
Abstract
In the past decade we have seen new advances and thus remarkable progress in the therapeutic options for non-small cell lung cancer (NSCLC). Among cytostatic therapies with new approaches in molecularly targeted therapies, we see new developments in a wide range of applications for immunotherapies. In this review we discuss the new potential modalities for the use of immune checkpoint inhibitors (ICIs) in the frontlines, including in early-stage (perioperative) and metastatic settings. The perioperative use of ICIs in both neoadjuvant and adjuvant settings may show benefits for patients. In early-stage NSCLC (from stage IIB and above) a multimodality approach is recommended as the gold standard for the treatment. After surgical resection platinum-based adjuvant chemotherapy has been the standard of care for many years. Based on the benefit of disease-free survival, the approval of adjuvant atezolizumab and adjuvant pembrolizumab was a significant breakthrough. In the metastatic setting, the use of immune checkpoint inhibitors with chemotherapy, regardless of PD-L1 expression or ICI alone (PD-L1 expression equal to or greater than 50%) also improves overall survival and progression-free survival.
Collapse
Affiliation(s)
- Attila Lieber
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Attila Makai
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Orosz
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Tamás Kardos
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Susil Joe Isaac
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Ilona Tornyi
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Nóra Bittner
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| |
Collapse
|
40
|
Meng Y, Zhou W, Li C, Zhou X, Li X, Li L, Fu Q, Huang J, Yue Y, Shen X, Yang L, Wang M. Case report: Response to tepotinib in Chinese non-small cell lung cancer patients harboring METex14 skipping with varying features. Front Oncol 2024; 14:1383964. [PMID: 39015492 PMCID: PMC11250067 DOI: 10.3389/fonc.2024.1383964] [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: 02/08/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024] Open
Abstract
MET exon 14 (METex14) skipping is the most reported MET mutation in non-small cell lung cancer (NSCLC) and has been confirmed to respond to MET tyrosine kinase inhibitors (TKI) in clinical trials. While MET TKI tepotinib was recently approved for METex14 skipping NSCLC in China, real-world evidence is limited. We report our experience treating NSCLC patients referred from oncology sites across China with tepotinib in the Boao Lecheng Pilot Zone. Four patients have been prescribed the drug with a median age of 67 years (range, 61-71 years). One patient has concomitant BRAF V600E mutation, and another patient had savolitinib as first line of therapy but discontinued due to hepatotoxicity. Till the end of follow-up, four patients were all on tepotinib therapy, with a median duration of therapy of 19 months. One patient achieved partial response and three achieved stable disease. Three patients had peripheral edema, but all were mild. Our experience showed in real clinical setting, tepotinib had robust and durable clinical activity and a favorable toxicity profile in Chinese patients with METex14 skipping NSCLC. It is the first report on the effectiveness of tepotinib in a patient with both METex14 skipping and BRAF V600E mutations and successful MET inhibitor switch after MET inhibitor-induced liver injury.
Collapse
Affiliation(s)
- Yan Meng
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Weiping Zhou
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Chenping Li
- Department of Pharmacy, Boao Super Hospital, Qionghai, Hainan, China
| | - Xiangjie Zhou
- Department of Pharmacy, Boao Super Hospital, Qionghai, Hainan, China
| | - Xiao Li
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Liang Li
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Qiye Fu
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Jue Huang
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Yali Yue
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Xuguang Shen
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Lijing Yang
- Department of Pharmacy, Boao Super Hospital, Qionghai, Hainan, China
| | - Meiqing Wang
- Department of Oncology II, Hainan Cancer Hospital, Haikou, Hainan, China
| |
Collapse
|
41
|
Mosele MF, Westphalen CB, Stenzinger A, Barlesi F, Bayle A, Bièche I, Bonastre J, Castro E, Dienstmann R, Krämer A, Czarnecka AM, Meric-Bernstam F, Michiels S, Miller R, Normanno N, Reis-Filho J, Remon J, Robson M, Rouleau E, Scarpa A, Serrano C, Mateo J, André F. Recommendations for the use of next-generation sequencing (NGS) for patients with advanced cancer in 2024: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2024; 35:588-606. [PMID: 38834388 DOI: 10.1016/j.annonc.2024.04.005] [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: 02/20/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Advancements in the field of precision medicine have prompted the European Society for Medical Oncology (ESMO) Precision Medicine Working Group to update the recommendations for the use of tumour next-generation sequencing (NGS) for patients with advanced cancers in routine practice. METHODS The group discussed the clinical impact of tumour NGS in guiding treatment decision using the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) considering cost-effectiveness and accessibility. RESULTS As for 2020 recommendations, ESMO recommends running tumour NGS in advanced non-squamous non-small-cell lung cancer, prostate cancer, colorectal cancer, cholangiocarcinoma, and ovarian cancer. Moreover, it is recommended to carry out tumour NGS in clinical research centres and under specific circumstances discussed with patients. In this updated report, the consensus within the group has led to an expansion of the recommendations to encompass patients with advanced breast cancer and rare tumours such as gastrointestinal stromal tumours, sarcoma, thyroid cancer, and cancer of unknown primary. Finally, ESMO recommends carrying out tumour NGS to detect tumour-agnostic alterations in patients with metastatic cancers where access to matched therapies is available. CONCLUSION Tumour NGS is increasingly expanding its scope and application within oncology with the aim of enhancing the efficacy of precision medicine for patients with cancer.
Collapse
Affiliation(s)
- M F Mosele
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich
| | - A Stenzinger
- Institute of Pathology, University Hospital Heidelberg and Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - F Barlesi
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre
| | - A Bayle
- Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre; Drug Development Department (DITEP), Gustave Roussy, Villejuif; Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - I Bièche
- Department of Genetics, Institut Curie, INSERM U1016, Université Paris Cité, Paris, France
| | - J Bonastre
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona; University of Vic-Central University of Catalonia, Vic, Spain; Oncoclínicas, São Paulo, Brazil
| | - A Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - A M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw; Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - R Miller
- Department of Medical Oncology, University College London, London; Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - N Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - J Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - J Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - M Robson
- Breast Medicine and Clinical Genetics Services, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Rouleau
- Tumor Genetics Service, Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
| | - A Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona-School of Medicine, Verona, Italy
| | - C Serrano
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - F André
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre.
| |
Collapse
|
42
|
Miao L, Qiu T, Li Y, Li J, Jiang X, Liu M, Zhang X, Jiang J, Zhang H, Wang Y, Li X, Ying J, Li M. Predicting MET exon 14 skipping mutation in pulmonary sarcomatoid carcinoma by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography features. Transl Lung Cancer Res 2024; 13:1232-1246. [PMID: 38973946 PMCID: PMC11225047 DOI: 10.21037/tlcr-24-56] [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: 01/16/2024] [Accepted: 05/07/2024] [Indexed: 07/09/2024]
Abstract
Background Pulmonary sarcomatoid carcinoma (PSC) is a rare, highly malignant type of non-small cell lung cancer (NSCLC) with a poor prognosis. Targeted drugs for MET exon 14 (METex14) skipping mutation can have considerable clinical benefits. This study aimed to predict METex14 skipping mutation in PSC patients by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography (CECT) features. Methods This retrospective study included 56 patients with PSC diagnosed by pathology. All patients underwent CECT before surgery or other treatment, and both targeted DNA- and RNA-based next-generation sequencing (NGS) were used to detect METex14 skipping mutation status. The patients were divided into two groups: METex14 skipping mutation and nonmutation groups. Overall, 1,316 texture features of the whole tumour were extracted. We also collected 12 clinical and 20 conventional CECT features. After dimensionality reduction and selection, predictive models were established by multivariate logistic regression analysis. Models were evaluated using the area under the curve (AUC), and the clinical utility of the model was assessed by decision curve analysis. Results METex14 skipping mutation was detected in 17.9% of PSCs. Mutations were found more frequently in those (I) who had smaller long- or short-axis diameters (P=0.02, P=0.01); (II) who had lower T stages (I, II) (P=0.02); and (III) with pseudocapsular or annular enhancement (P=0.03). The combined model based on the conventional and texture models yielded the best performance in predicting METex14 skipping mutation with the highest AUC (0.89). The conventional and texture models also had good performance (AUC =0.83 conventional; =0.88 texture). Conclusions Whole-tumour texture analysis combined with clinical and conventional CECT features may serve as a noninvasive tool to predict the METex14 skipping mutation status in PSC.
Collapse
Affiliation(s)
- Lei Miao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Li
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Jiang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengwen Liu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiuming Jiang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huanhuan Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmei Wang
- GE Healthcare China, Pudong New Area, Shanghai, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Li
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Imyanitov EN, Preobrazhenskaya EV, Orlov SV. Current status of molecular diagnostics for lung cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:742-765. [PMID: 38966170 PMCID: PMC11220319 DOI: 10.37349/etat.2024.00244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/08/2024] [Indexed: 07/06/2024] Open
Abstract
The management of lung cancer (LC) requires the analysis of a diverse spectrum of molecular targets, including kinase activating mutations in EGFR, ERBB2 (HER2), BRAF and MET oncogenes, KRAS G12C substitutions, and ALK, ROS1, RET and NTRK1-3 gene fusions. Administration of immune checkpoint inhibitors (ICIs) is based on the immunohistochemical (IHC) analysis of PD-L1 expression and determination of tumor mutation burden (TMB). Clinical characteristics of the patients, particularly age, gender and smoking history, significantly influence the probability of finding the above targets: for example, LC in young patients is characterized by high frequency of kinase gene rearrangements, while heavy smokers often have KRAS G12C mutations and/or high TMB. Proper selection of first-line therapy influences overall treatment outcomes, therefore, the majority of these tests need to be completed within no more than 10 working days. Activating events in MAPK signaling pathway are mutually exclusive, hence, fast single-gene testing remains an option for some laboratories. RNA next-generation sequencing (NGS) is capable of detecting the entire repertoire of druggable gene alterations, therefore it is gradually becoming a dominating technology in LC molecular diagnosis.
Collapse
Affiliation(s)
- Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Clinical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
- I.V. Kurchatov Complex for Medical Primatology, National Research Centre “Kurchatov Institute”, 354376 Sochi, Russia
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Clinical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Sergey V. Orlov
- I.V. Kurchatov Complex for Medical Primatology, National Research Centre “Kurchatov Institute”, 354376 Sochi, Russia
- Department of Oncology, I.P. Pavlov St.-Petersburg State Medical University, 197022 St.-Petersburg, Russia
| |
Collapse
|
44
|
Wang X, Bai L, Kong L, Guo Z. Advances in circulating tumor cells for early detection, prognosis and metastasis reduction in lung cancer. Front Oncol 2024; 14:1411731. [PMID: 38974237 PMCID: PMC11224453 DOI: 10.3389/fonc.2024.1411731] [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: 04/03/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Globally, lung cancer stands as the leading type of cancer in terms of incidence and is the major source of mortality attributed to cancer. We have outlined the molecular biomarkers for lung cancer that are available clinically. Circulating tumor cells (CTCs) spread from the original location, circulate in the bloodstream, extravasate, and metastasize, forming secondary tumors by invading and establishing a favorable environment. CTC analysis is considered a common liquid biopsy method for lung cancer. We have enumerated both in vivo and ex vivo techniques for CTC separation and enrichment, examined the advantages and limitations of these methods, and also discussed the detection of CTCs in other bodily fluids. We have evaluated the value of CTCs, as well as CTCs in conjunction with other biomarkers, for their utility in the early detection and prognostic assessment of patients with lung cancer. CTCs engage with diverse cells of the metastatic process, interfering with the interaction between CTCs and various cells in metastasis, potentially halting metastasis and enhancing patient prognosis.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Lu Bai
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Linghui Kong
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Zhijuan Guo
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
45
|
Mohan CD, Shanmugam MK, Gowda SGS, Chinnathambi A, Rangappa KS, Sethi G. c-MET pathway in human malignancies and its targeting by natural compounds for cancer therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155379. [PMID: 38503157 DOI: 10.1016/j.phymed.2024.155379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND c-MET is a receptor tyrosine kinase which is classically activated by HGF to activate its downstream signaling cascades such as MAPK, PI3K/Akt/mTOR, and STAT3. The c-MET modulates cell proliferation, epithelial-mesenchymal transition (EMT), immune response, morphogenesis, apoptosis, and angiogenesis. The c-MET has been shown to serve a prominent role in embryogenesis and early development. The c-MET pathway is deregulated in a broad range of malignancies, due to overexpression of ligands or receptors, genomic amplification, and MET mutations. The link between the deregulation of c-MET signaling and tumor progression has been well-documented. Overexpression or overactivation of c-MET is associated with dismal clinical outcomes and acquired resistance to targeted therapies. Since c-MET activation results in the triggering of oncogenic pathways, abrogating the c-MET pathway is considered to be a pivotal strategy in cancer therapeutics. Herein, an analysis of role of the c-MET pathway in human cancers and its relevance in bone metastasis and therapeutic resistance has been undertaken. Also, an attempt has been made to summarize the inhibitory activity of selected natural compounds towards c-MET signaling in cancers. METHODS The publications related to c-MET pathway in malignancies and its natural compound modulators were obtained from databases such as PubMed, Scopus, and Google Scholar and summarized based on PRISMA guidelines. Some of the keywords used for extracting relevant literature are c-MET, natural compound inhibitors of c-MET, c-MET in liver cancer, c-MET in breast cancer, c-MET in lung cancer, c-MET in pancreatic cancer, c-MET in head and neck cancer, c-MET in bone metastasis, c-MET in therapeutic resistance, and combination of c-MET inhibitors and chemotherapeutic agents. The chemical structure of natural compounds was verified in PubChem database. RESULTS The search yielded 3935 publications, of which 195 reference publications were used for our analysis. Clinical trials were referenced using ClinicalTrials.gov identifier. The c-MET pathway has been recognized as a prominent target to combat the growth, metastasis, and chemotherapeutic resistance in cancers. The key role of the c-MET in bone metastasis as well as therapeutic resistance has been elaborated. Also, suppressive effect of selected natural compounds on the c-MET pathway in clinical/preclinical studies has been discussed.
Collapse
Affiliation(s)
- Chakrabhavi Dhananjaya Mohan
- FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226 001, India
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | | | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Kanchugarakoppal S Rangappa
- Institution of Excellence, Vijnana Bhavan, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
46
|
Borczuk AC. Molecular Testing in Lung Cancer: Recommendations and Update. Surg Pathol Clin 2024; 17:307-320. [PMID: 38692813 DOI: 10.1016/j.path.2023.11.012] [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] [Indexed: 05/03/2024]
Abstract
Adoption of molecular testing in lung cancer is increasing. Molecular testing for staging and prediction of response for targeted therapy remain the main indications, and although utilization of blood-based testing for tumor is growing, the use of the diagnostic cytology and tissue specimens is equally important. The pathologist needs to optimize reflex testing, incorporate stage-based algorithms, and understand types of tests for timely and complete assessment in the majority of cases. When tissue is limited, testing should capture the most frequent alterations to maximize the yield of what are largely mutually exclusive alterations, avoiding the need for repeat biopsy.
Collapse
Affiliation(s)
- Alain C Borczuk
- Anatomic Pathology, Northwell Health, 2200 Northern Boulevard Suite 104, Greenvale, NY 11548, USA.
| |
Collapse
|
47
|
Dias e Silva D, Mambetsariev I, Fricke J, Babikian R, Dingal ST, Mazdisnian F, Badie B, Arvanitis L, Afkhami M, Villalona-Calero M, Salgia R. A novel HLA-DQB2::MET gene fusion variant in lung adenocarcinoma with prolonged response to tepotinib: a case report. Transl Lung Cancer Res 2024; 13:1163-1168. [PMID: 38854944 PMCID: PMC11157375 DOI: 10.21037/tlcr-24-34] [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: 01/11/2024] [Accepted: 03/20/2024] [Indexed: 06/11/2024]
Abstract
Background MET rearrangements are infrequently observed in non-small cell lung cancer (NSCLC). Advanced genomic detection techniques have unveiled such infrequent genomic variations, particularly MET fusions in approximately 0.5% of NSCLC patients. Tyrosine kinase inhibitors (TKIs) have revolutionized the standard of care in lung cancer and more recently a second generation MET TKI tepotinib received Food and Drug Administration (FDA) approval for MET exon 14 alterations in metastatic NSCLC. Despite this, the therapeutic landscape for MET-rearranged NSCLC patients remains significantly unexplored. The aim of our report is to detail a unique case of a patient with metastatic lung adenocarcinoma with a novel HLA-DQB2::MET fusion detected by next-generation sequencing (NGS) following previous treatment resistance. Case Description A 73-year-old female was initially started on carboplatin, pemetrexed and pembrolizumab with maintenance, but eventually had progression in the left upper lobe (LUL). Upon progression she was enrolled in a clinical trial of a monoclonal antibody with or without a PD-1 inhibitor, but brain metastasis progression was eventually detected by magnetic resonance imaging (MRI) requiring stereotactic radiosurgery (SRS) and a craniotomy. The trial drug was eventually discontinued due to progression and toxicity and NGS on bronchoscopy tissue revealed HLA-DQB2::MET fusion. The patient was initiated on tepotinib and continues with clinical and radiological stable disease for over 12 months. The patient's response to a MET inhibitor, tepotinib, underscores the potential efficacy of selective MET inhibitors for individuals with previously unexplored MET fusions. Conclusions The positive response to tepotinib of a patient with NSCLC harboring a novel MET-Fusion underscores the importance of the use of comprehensive next-generational sequencing-based panels and highlights the necessity for additional research and clinical exploration of selective MET inhibitors for managing NSCLC with MET rearrangements.
Collapse
Affiliation(s)
- Douglas Dias e Silva
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Razmig Babikian
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Shaira Therese Dingal
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Behnam Badie
- Department of Surgery, City of Hope, Duarte, CA, USA
| | | | | | - Miguel Villalona-Calero
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
48
|
Thu YM, Suzawa K, Tomida S, Ochi K, Tsudaka S, Takatsu F, Date K, Matsuda N, Iwata K, Nakata K, Shien K, Yamamoto H, Okazaki M, Sugimoto S, Toyooka S. PAI-1 mediates acquired resistance to MET-targeted therapy in non-small cell lung cancer. PLoS One 2024; 19:e0300644. [PMID: 38758826 PMCID: PMC11101109 DOI: 10.1371/journal.pone.0300644] [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: 12/20/2023] [Accepted: 03/03/2024] [Indexed: 05/19/2024] Open
Abstract
Mechanisms underlying primary and acquired resistance to MET tyrosine kinase inhibitors (TKIs) in managing non-small cell lung cancer remain unclear. In this study, we investigated the possible mechanisms acquired for crizotinib in MET-amplified lung carcinoma cell lines. Two MET-amplified lung cancer cell lines, EBC-1 and H1993, were established for acquired resistance to MET-TKI crizotinib and were functionally elucidated. Genomic and transcriptomic data were used to assess the factors contributing to the resistance mechanism, and the alterations hypothesized to confer resistance were validated. Multiple mechanisms underlie acquired resistance to crizotinib in MET-amplified lung cancer cell lines. In EBC-1-derived resistant cells, the overexpression of SERPINE1, the gene encoding plasminogen activator inhibitor-1 (PAI-1), mediated the drug resistance mechanism. Crizotinib resistance was addressed by combination therapy with a PAI-1 inhibitor and PAI-1 knockdown. Another mechanism of resistance in different subline cells of EBC-1 was evaluated as epithelial-to-mesenchymal transition with the upregulation of antiapoptotic proteins. In H1993-derived resistant cells, MEK inhibitors could be a potential therapeutic strategy for overcoming resistance with downstream mitogen-activated protein kinase pathway activation. In this study, we revealed the different mechanisms of acquired resistance to the MET inhibitor crizotinib with potential therapeutic application in patients with MET-amplified lung carcinoma.
Collapse
Affiliation(s)
- Yin Min Thu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ken Suzawa
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Kosuke Ochi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shimpei Tsudaka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fumiaki Takatsu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keiichi Date
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Naoki Matsuda
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuma Iwata
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Nakata
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Kazuhiko Shien
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Yamamoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
49
|
Wang Y, Xu M, Wang K, Hao Y, Xu C, Song Z. Assessment of efficacy and safety of MET tyrosine kinase inhibitors in non-small-cell lung cancer patients with MET alterations. Ther Adv Med Oncol 2024; 16:17588359241248352. [PMID: 38736555 PMCID: PMC11088810 DOI: 10.1177/17588359241248352] [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: 12/21/2023] [Accepted: 04/03/2024] [Indexed: 05/14/2024] Open
Abstract
Background While targeted therapy has become the standard treatment for certain non-small-cell lung cancer (NSCLC) patients with gene mutation positivity, there remains a lack of enough reports of the efficacy of mesenchymal-epithelial transition (MET) alterations in the real world. Objectives We aimed to explore the efficacy and toxicity of targeted therapy in NSCLC patients with different types of MET alterations and hope to provide more clinical medication guidance. Design Designed different subgroups to compare the efficacy and safety of targeted therapy in NSCLC patients with MET alterations. Methods We conducted analyses on the efficacy and safety of mesenchymal-epithelial transition factor-tyrosine kinase inhibitor (MET-TKI) therapy in NSCLC patients with MET alterations. Tumor response was evaluated based on the Response Evaluation Criteria in Solid Tumors version 1.1 criteria, and both progression-free survival (PFS) and overall survival were determined using the Kaplan-Meier method. Results Our study encompassed 116 NSCLC patients with MET alterations, including MET ex14 skipping mutation (n = 50), MET primary amplification (amp) (n = 25), and secondary amp (n = 41). Among treated patients, 34 achieved a partial response, while 52 exhibited stable disease. The overall response rate for the entire cohort was 29.31%, with a disease control rate of 74.14%. A significant difference was observed in the median PFS among patients with MET ex14 skipping mutation, MET primary amplification (amp), and secondary amp (10.4 versus 6.6 versus 4.5 months, p = 0.002). In all, 69 patients experienced drug-related adverse effects, with the most common being peripheral edema (35.34%), nausea and vomiting (21.55%), and fatigue (10.34%). In total, 29 patients (25%) encountered drug-related adverse reactions of grade 3 or higher. Conclusion MET-TKI therapy works better for MET ex14 skipping mutation than other types of MET gene alteration. In the two MET amplified groups, the secondary amp was less effective. This study may provide more research support for the treatment of these patients.
Collapse
Affiliation(s)
- Yanhua Wang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Manyi Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ke Wang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yue Hao
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Chunwei Xu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Zhengbo Song
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu, Hangzhou, Zhejiang 310022, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
50
|
Gou Q, Gou Q, Gan X, Xie Y. Novel therapeutic strategies for rare mutations in non-small cell lung cancer. Sci Rep 2024; 14:10317. [PMID: 38705930 PMCID: PMC11070427 DOI: 10.1038/s41598-024-61087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/30/2024] [Indexed: 05/07/2024] Open
Abstract
Lung cancer is still the leading cause of cancer-related mortality. Over the past two decades, the management of non-small cell lung cancer (NSCLC) has undergone a significant revolution. Since the first identification of activating mutations in the epidermal growth factor receptor (EGFR) gene in 2004, several genetic aberrations, such as anaplastic lymphoma kinase rearrangements (ALK), neurotrophic tropomyosin receptor kinase (NTRK) and hepatocyte growth factor receptor (MET), have been found. With the development of gene sequencing technology, the development of targeted drugs for rare mutations, such as multikinase inhibitors, has provided new strategies for treating lung cancer patients with rare mutations. Patients who harbor this type of oncologic driver might acquire a greater survival benefit from the use of targeted therapy than from the use of chemotherapy and immunotherapy. To date, more new agents and regimens can achieve satisfactory results in patients with NSCLC. In this review, we focus on recent advances and highlight the new approval of molecular targeted therapy for NSCLC patients with rare oncologic drivers.
Collapse
Affiliation(s)
- Qitao Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiheng Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiaochuan Gan
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxin Xie
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|