1
|
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
|
2
|
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
|
3
|
Yu QX, Fu PY, Zhang C, Li L, Huang WT. Mesenchymal-epithelial transition factor amplification correlates with adverse pathological features and poor clinical outcome in colorectal cancer. World J Gastrointest Surg 2024; 16:1395-1406. [PMID: 38817281 PMCID: PMC11135301 DOI: 10.4240/wjgs.v16.i5.1395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/22/2024] [Accepted: 04/11/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer and the second most common cause of cancer-related mortality worldwide. Mesenchymal-epithelial transition factor (MET) gene participates in multiple tumor biology and shows clinical potential for pharmacological manipulation in tumor treatment. MET amplification has been reported in CRC, but data are very limited. Investigating pathological values of MET in CRC may provide new therapeutic and genetic screening options in future clinical practice. AIM To determine the pathological significance of MET amplification in CRC and to propose a feasible screening strategy. METHODS A number of 205 newly diagnosed CRC patients undergoing surgical resection without any preoperative therapy at Shenzhen Cancer Hospital of Chinese Academy of Medical Sciences were recruited. All patients were without RAS/RAF mutation or microsatellite instability-high. MET amplification and c-MET protein expression were analyzed using fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC), respectively. Correlations between MET aberration and pathological features were detected using the chi-squared test. Progression free survival (PFS) during the two-year follow-up was detected using the Kaplan-Meier method and log rank test. The results of MET FISH and IHC were compared using one-way ANOVA. RESULTS Polysomy-induced MET amplification was observed in 14.4% of cases, and focal MET amplification was not detected. Polysomy-induced MET amplification was associated with a higher frequency of lymph node metastasis (LNM) (P < 0.001) and higher tumor budding grade (P = 0.02). In the survival analysis, significant difference was detected between patients with amplified- and non-amplified MET in a two-year follow-up after the first diagnosis (P = 0.001). C-MET scores of 0, 1+, 2+, and 3+ were observed in 1.4%, 24.9%, 54.7%, and 19.0% of tumors, respectively. C-MET overexpression correlated with higher frequency of LNM (P = 0.002), but no significant difference of PFS was detected between patients with different protein levels. In terms of concordance between MET FISH and IHC results, MET copy number showed no difference in c-MET IHC 0/1+ (3.35 ± 0.18), 2+ (3.29 ± 0.11) and 3+ (3.58 ± 0.22) cohorts, and the MET-to-CEP7 ratio showed no difference in three groups (1.09 ± 0.02, 1.10 ± 0.01, and 1.09 ± 0.03). CONCLUSION In CRC, focal MET amplification was a rare event. Polysomy-induced MET amplification correlated with adverse pathological characteristics and poor prognosis. IHC was a poor screening tool for MET amplification.
Collapse
Affiliation(s)
- Qiu-Xiao Yu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, Guangdong Province, China
| | - Ping-Ying Fu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, Guangdong Province, China
| | - Chi Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, Guangdong Province, China
| | - Li Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, Guangdong Province, China
| | - Wen-Ting Huang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, Guangdong Province, China
| |
Collapse
|
4
|
Xiang C, Lv X, Chen K, Guo L, Zhao R, Teng H, Ye M, Kuang T, Hou T, Liu C, Du H, Zhang Z, Han Y. Unraveling the Significance of MET Focal Amplification in Lung Cancer: Integrative NGS, FISH, and IHC Investigation. Mod Pathol 2024; 37:100451. [PMID: 38369190 DOI: 10.1016/j.modpat.2024.100451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/24/2024] [Accepted: 02/04/2024] [Indexed: 02/20/2024]
Abstract
MET amplification (METamp) represents a promising therapeutic target in non-small cell lung cancer, but no consensus has been established to identify METamp-dependent tumors that could potentially benefit from MET inhibitors. In this study, an analysis of MET amplification/overexpression status was performed in a retrospectively recruited cohort comprising 231 patients with non-small cell lung cancer from Shanghai Chest Hospital (SCH cohort) using 3 methods: fluorescence in situ hybridization (FISH), hybrid capture-based next-generation sequencing, and immunohistochemistry for c-MET and phospho-MET. The SCH cohort included 130 cases known to be METamp positive by FISH and 101 negative controls. The clinical relevance of these approaches in predicting the efficacy of MET inhibitors was evaluated. Additionally, next-generation sequencing data from another 2 cohorts including 22,010 lung cancer cases were utilized to examine the biological characteristics of different METamp subtypes. Of the 231 cases, 145 showed MET amplification/overexpression using at least 1 method, whereas only half of them could be identified by all 3 methods. METamp can occur as focal amplification or polysomy. Our study revealed that the inconsistency between next-generation sequencing and FISH primarily occurred in the polysomy subtype. Further investigations indicated that compared with polysomy, focal amplification correlated with fewer co-occurring driver mutations, higher protein expressions of c-MET and phospho-MET, and higher incidence in acquired resistance than in de novo setting. Moreover, patients with focal amplification presented a more robust response to MET inhibitors compared with those with polysomy. Notably, a strong correlation was observed between focal amplification and programmed cell death ligand-1 expression, indicating potential therapeutic implications with combined MET inhibitor and immunotherapy for patients with both alterations. Our findings provide insights into the molecular complexity and clinical relevance of METamp in lung cancer, highlighting the role of MET focal amplification as an oncogenic driver and its feasibility as a primary biomarker to further investigate the clinical activity of MET inhibitors in future studies.
Collapse
Affiliation(s)
- Chan Xiang
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinze Lv
- Burning Rock Biotech, Guangzhou, China
| | - Ke Chen
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lianying Guo
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiying Zhao
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haohua Teng
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Ye
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Ting Hou
- Burning Rock Biotech, Guangzhou, China
| | | | - Haiwei Du
- Burning Rock Biotech, Guangzhou, China
| | | | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Han Y, Yu Y, Miao D, Zhou M, Zhao J, Shao Z, Jin R, Le X, Li W, Xia Y. Targeting MET in NSCLC: An Ever-Expanding Territory. JTO Clin Res Rep 2024; 5:100630. [PMID: 38361739 PMCID: PMC10867448 DOI: 10.1016/j.jtocrr.2023.100630] [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: 09/02/2023] [Revised: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 02/17/2024] Open
Abstract
MET protooncogene (MET) alterations are known driver oncogenes in NSCLC. Since the identification of MET as a potential therapeutic target, extensive clinical trials have been performed. As a result, MET-targeted therapies, including MET tyrosine kinase inhibitors, monoclonal antibodies, and MET antibody-drug conjugates now play important roles in the standard treatment of MET-altered NSCLC; they have considerably improved the outcomes of patients with tumors that harbor MET oncogenic drivers. Although clinical agents are currently available and numerous other options are in development, particular challenges in the field require attention. For example, the therapeutic efficacy of each drug remains unsatisfactory, and concomitantly, the resistance mechanisms are not fully understood. Thus, there is an urgent need for optimal drug sequencing and combinations, along with a thorough understanding of treatment resistance. In this review, we describe the current landscape of pertinent clinical trials focusing on MET-targeted strategies and discuss future developmental directions in this rapidly expanding field.
Collapse
Affiliation(s)
- Ying Han
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Yinghui Yu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Da Miao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Mo Zhou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Jing Zhao
- Department of Medical Oncology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Zhehua Shao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Rui Jin
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - 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, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of 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, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
6
|
Yuan P, Xue X, Qiu T, Ying J. MET alterations detection platforms and clinical implications in solid tumors: a comprehensive review of literature. Ther Adv Med Oncol 2024; 16:17588359231221910. [PMID: 38249331 PMCID: PMC10798113 DOI: 10.1177/17588359231221910] [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: 06/02/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
MET alterations, including MET exon 14 skipping variants, MET amplification, MET overexpression, and MET fusion, play pivotal roles in primary tumorigenesis and acquired resistance to targeted therapies, especially EGFR tyrosine kinase inhibitors. They represent important diagnostic, prognostic, and predictive biomarkers in many solid tumor types. However, the detection of MET alterations is challenging due to the complexity of MET alterations and the diversity of platform technologies. Therefore, techniques with high sensitivity, specificity, and reliable molecular detection accuracy are needed to overcome such hindrances and aid in biomarker-guided therapies. The current review emphasizes the role of MET alterations as oncogenic drivers in a variety of cancers and their involvement in the development of resistance to targeted therapies. Moreover, our review provides an overview of and recommendations on the selection of various cross-platform technologies for the detection of MET exon 14 skipping variants, MET amplification, MET overexpression, and MET fusion. Furthermore, challenges and hurdles underlying these common detection platforms are discussed.
Collapse
Affiliation(s)
- Pei Yuan
- Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuemin Xue
- Department of Pathology, State Key Laboratory of Molecular Oncology, 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, State Key Laboratory of Molecular Oncology, 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, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| |
Collapse
|
7
|
Bontoux C, Hofman V, Abboute M, Lespinet-Fabre V, Lalvée S, Goffinet S, Bordone O, Long-Mira E, Lassalle S, Murcy F, Rignol G, Heeke S, Ilie M, Hofman P. c-Met immunohistochemistry as reflex test at diagnosis for non-small cell lung cancer: a real-world experience from a monocentric case series. J Clin Pathol 2023:jcp-2023-209202. [PMID: 37940375 DOI: 10.1136/jcp-2023-209202] [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: 09/19/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023]
Abstract
AIMS Recent clinical trials have shown promising results with drugs targeting the hepatocyte growth factor receptor (c-Met) for advanced non-small cell lung cancers overexpressing c-Met. We assessed reflex testing of c-Met immunohistochemistry (IHC) at diagnosis for NSCLC in the real-world. METHODS We retrospectively collected clinical, pathological and molecular data of cases diagnosed with NSCLC in our institution from January 2021 to June 2023. We performed c-Met IHC (SP44 clone) and scored the expression using a H-score and a three-tier classification. RESULTS 391 cases with interpretable c-Met IHC staining were included. The median age at diagnosis was 70 years (range 25-89 years) including 234 males (male/female ratio 1:5). 58% of the samples came from surgical resections, 35% from biopsies and 8% from cytological procedures. 52% of cases were classified as c-Met-positive (H-score≥150) and 19% were classified as c-Methigh (≥50%, 3+). 43% of the c-Metneg presented with lymph node and/or visceral metastases at diagnosis vs 55% for c-Methigh (p=0.042). 23% of the adenocarcinomas showed c-Methigh expression vs 3% for squamous cell carcinomas (p=0.004). 27% of the c-Metneg cases had a high PD-L1 expression vs 58% of c-Methigh cases (p<0.001). MET ex14 skipping was present in 8% of the c-Methigh cases. CONCLUSIONS Systematic c-Met testing in daily routine for NSCLC patients is feasible, highlighting a potential correlation with clinicopathological and molecular features.
Collapse
Affiliation(s)
- Christophe Bontoux
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
| | - Veronique Hofman
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Milissa Abboute
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Virginie Lespinet-Fabre
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Salomé Lalvée
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Samantha Goffinet
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Olivier Bordone
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Elodie Long-Mira
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Sandra Lassalle
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Florent Murcy
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Guylène Rignol
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marius Ilie
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Paul Hofman
- Team 4, Inserm U1081, CNRS 7284, Université Côte d'Azur, Antoine Lacassagne Cancer Center, IRCAN, Nice, France
- IHU RespirERA, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Centre Nice Laboratory of Clinical and Experimental Pathology, Nice, France
| |
Collapse
|
8
|
Fernandes M, Hoggard B, Jamme P, Paget S, Truong M, Grégoire V, Vinchent A, Descarpentries C, Morabito A, Stanislovas J, Farage E, Meneboo J, Sebda S, Bouchekioua‐Bouzaghou K, Nollet M, Humez S, Perera T, Fromme P, Grumolato L, Figeac M, Copin M, Tulasne D, Cortot AB, Kermorgant S, Kherrouche Z. MET exon 14 skipping mutation is a hepatocyte growth factor (HGF)-dependent oncogenic driver in vitro and in humanised HGF knock-in mice. Mol Oncol 2023; 17:2257-2274. [PMID: 36799689 PMCID: PMC10620121 DOI: 10.1002/1878-0261.13397] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/27/2022] [Accepted: 02/16/2023] [Indexed: 02/18/2023] Open
Abstract
Exon skipping mutations of the MET receptor tyrosine kinase (METex14), increasingly reported in cancers, occur in 3-4% of non-small-cell lung cancer (NSCLC). Only 50% of patients have a beneficial response to treatment with MET-tyrosine kinase inhibitors (TKIs), underlying the need to understand the mechanism of METex14 oncogenicity and sensitivity to TKIs. Whether METex14 is a driver mutation and whether it requires hepatocyte growth factor (HGF) for its oncogenicity in a range of in vitro functions and in vivo has not been fully elucidated from previous preclinical models. Using CRISPR/Cas9, we developed a METex14/WT isogenic model in nontransformed human lung cells and report that the METex14 single alteration was sufficient to drive MET-dependent in vitro anchorage-independent survival and motility and in vivo tumorigenesis, sensitising tumours to MET-TKIs. However, we also show that human HGF (hHGF) is required, as demonstrated in vivo using a humanised HGF knock-in strain of mice and further detected in tumour cells of METex14 NSCLC patient samples. Our results also suggest that METex14 oncogenicity is not a consequence of an escape from degradation in our cell model. Thus, we developed a valuable model for preclinical studies and present results that have potential clinical implication.
Collapse
Affiliation(s)
- Marie Fernandes
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | | | - Philippe Jamme
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | - Sonia Paget
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | - Marie‐José Truong
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | | | - Audrey Vinchent
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | | | - Angela Morabito
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | | | - Enoir Farage
- Barts Cancer InstituteQueen Mary University of LondonUK
| | - Jean‐Pascal Meneboo
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, LilleFrance
| | - Shéhérazade Sebda
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, LilleFrance
| | | | - Marie Nollet
- Barts Cancer InstituteQueen Mary University of LondonUK
| | - Sarah Humez
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
- Univ LilleDepartment of Pathology, CHU LilleFrance
| | | | - Paul Fromme
- Department of Mechanical EngineeringUniversity College LondonUK
| | - Luca Grumolato
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, 76000 RouenFrance
| | - Martin Figeac
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, LilleFrance
| | - Marie‐Christine Copin
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
- Univ LilleDepartment of Pathology, CHU LilleFrance
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| | - Alexis B. Cortot
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
- Univ. LilleThoracic Oncology Department, CHU LilleFrance
| | | | - Zoulika Kherrouche
- Univ. Lille, CNRS, Inserm, CHU LilleInstitut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesFrance
| |
Collapse
|
9
|
Kumaki Y, Oda G, Ikeda S. Targeting MET Amplification: Opportunities and Obstacles in Therapeutic Approaches. Cancers (Basel) 2023; 15:4552. [PMID: 37760522 PMCID: PMC10526812 DOI: 10.3390/cancers15184552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
The MET gene plays a vital role in cellular proliferation, earning it recognition as a principal oncogene. Therapies that target MET amplification have demonstrated promising results both in preclinical models and in specific clinical cases. A significant obstacle to these therapies is the ability to distinguish between focal amplification and polysomy, a task for which simple MET copy number measurement proves insufficient. To effectively differentiate between the two, it is crucial to utilize comparative measures, including in situ hybridization (ISH) with the centromere or next generation sequencing (NGS) with adjacent genes. Despite the promising potential of MET amplification treatment, the judicious selection of patients is paramount to maximize therapeutic efficacy. The effectiveness of MET inhibitors can fluctuate depending on the extent of MET amplification. Future research must seek to establish the ideal threshold value for MET amplification, identify the most efficacious combination therapies, and innovate new targeted treatments for patients exhibiting MET amplification.
Collapse
Affiliation(s)
- Yuichi Kumaki
- Department of Specialized Surgery, Tokyo Medical and Dental University, Tokyo 113-8519, Japan;
| | - Goshi Oda
- Department of Specialized Surgery, Tokyo Medical and Dental University, Tokyo 113-8519, Japan;
| | - Sadakatsu Ikeda
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
10
|
Mazieres J, Vioix H, Pfeiffer BM, Campden RI, Chen Z, Heeg B, Cortot AB. MET Exon 14 Skipping in NSCLC: A Systematic Literature Review of Epidemiology, Clinical Characteristics, and Outcomes. Clin Lung Cancer 2023; 24:483-497. [PMID: 37451931 DOI: 10.1016/j.cllc.2023.06.008] [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: 02/15/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION MET exon 14 (METex14) skipping is a rare oncogenic driver in non-small-cell lung cancer (NSCLC) for which targeted therapy with MET tyrosine kinase inhibitors (TKIs) was recently approved. Given the heterogeneity in published data of METex14 skipping NSCLC, we conducted a systematic literature review to evaluate its frequency, patient characteristics, and outcomes. METHODS On June 13, 2022 we conducted a systematic literature review of publications and conference abstracts reporting frequency, patient characteristics, or outcomes of patients with METex14 skipping NSCLC. RESULTS We included 139 studies reporting frequency or patient characteristics (350,997 patients), and 39 studies reporting clinical outcomes (3989 patients). Median METex14 skipping frequency was 2.0% in unselected patients with NSCLC, with minimal geographic variation. Median frequency was 2.4% in adenocarcinoma or nonsquamous subgroups, 12.0% in sarcomatoid, and 1.3% in squamous histology. Patients with METex14 skipping NSCLC were more likely to be elderly, have adenocarcinoma histology; there was no marked sex or smoking status distribution. In first line of treatment, median objective response rate ranged from 50.7% to 68.8% with targeted therapies (both values correspond to MET TKIs), was 33.3% with immunotherapy, and ranged from 23.1% to 27.0% with chemotherapy. CONCLUSIONS Patients with METex14 skipping are more likely to have certain characteristics, but no patient subgroup can be ruled out; thus, it is crucial to test all patients with NSCLC to identify suitable candidates for MET inhibitor therapy. MET TKIs appeared to result in higher efficacy outcomes, although no direct comparison with chemotherapy or immunotherapy regimens was found.
Collapse
Affiliation(s)
- Julien Mazieres
- CHU de Toulouse, Université Paul Sabatier, Toulouse, France.
| | - Helene Vioix
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | | | - Zhiyuan Chen
- Ingress Health, A Cytel Company, Rotterdam, The Netherlands
| | - Bart Heeg
- Ingress Health, A Cytel Company, Rotterdam, The Netherlands
| | - Alexis B Cortot
- Université Lille, Centre Hospitalier Universitaire de Lille, Centre national de la recherche scientifique, Inserm, Institute Pasteur de Lille, Lille, France
| |
Collapse
|
11
|
Urbanska EM, Grauslund M, Koffeldt PR, Truelsen SLB, Löfgren JO, Costa JC, Melchior LC, Sørensen JB, Santoni-Rugiu E. Real-World Data on Combined EGFR-TKI and Crizotinib Treatment for Acquired and De Novo MET Amplification in Patients with Metastatic EGFR-Mutated NSCLC. Int J Mol Sci 2023; 24:13077. [PMID: 37685884 PMCID: PMC10487649 DOI: 10.3390/ijms241713077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Amplification of the mesenchymal epithelial transition (MET) gene is a mechanism of acquired resistance to epidermal growth factor receptor (EGFR)-tyrosine-kinase-inhibitors (TKIs) in over 20% of patients with advanced EGFR-mutated (EGFRm+) non-small lung cancer (NSCLC). However, it may also occur de novo in 2-8% of EGFRm+ NSCLC cases as a potential mechanism of intrinsic resistance. These patients represent a group with unmet needs, since there is no standard therapy currently approved. Several new MET inhibitors are being investigated in clinical trials, but the results are awaited. Meanwhile, as an alternative strategy, combinations of EGFR-TKIs with the MET/ALK/ROS1-TKI Crizotinib may be used in this setting, despite this use is principally off-label. Thus, we studied five of these MET amplified cases receiving EGFR-TKI and Crizotinib doublet after progression on EGFR-TKI treatment to assess the benefits and challenges related to this combination and the possible occurrence of genomic and phenotypic co-alterations. Furthermore, we compared our cases with other real-world reports on Crizotinib/EGFR-TKI combinations, which appeared effective, especially in patients with high-level MET amplification. Yet, we observed that the co-occurrence of other genomic and phenotypical alterations may affect the response to combined EGFR-TKI and Crizotinib. Finally, given the heterogeneity of MET amplification, the diagnostic methods for assessing it may be discrepant. In this respect, we observed that for optimal detection, immunohistochemistry, fluorescence in situ hybridization, and next-generation sequencing should be used together, as these methods possess different sensitivities and complement each other in characterizing MET amplification. Additionally, we addressed the issue of managing EGFR-mutated NSCLC patients with de novo MET amplification causing primary EGFR-TKI resistance. We conclude that, while data from clinical trials with new MET inhibitors are still pending, adding Crizotinib to EGFR-TKI in NSCLC patients acquiring MET amplification at progression on EGFR-TKI monotherapy is a reasonable approach, with a progression-free survival of 3-19 months.
Collapse
Affiliation(s)
- Edyta M. Urbanska
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
| | - Morten Grauslund
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Peter R. Koffeldt
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Sarah L. B. Truelsen
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Johan O. Löfgren
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
| | - Junia C. Costa
- Department of Radiology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
| | - Linea C. Melchior
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Jens B. Sørensen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Eric Santoni-Rugiu
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
12
|
Gilson P, Pouget C, Belmonte R, Fadil S, Demange J, Rouyer M, Lacour J, Betz M, Dardare J, Witz A, Merlin JL, Harlé A. Validation of the Idylla GeneFusion assay to detect fusions and MET exon-skipping in non-small cell lung cancers. Sci Rep 2023; 13:12909. [PMID: 37558711 PMCID: PMC10412571 DOI: 10.1038/s41598-023-39749-4] [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: 03/07/2023] [Accepted: 07/30/2023] [Indexed: 08/11/2023] Open
Abstract
Gene fusions and MET exon skipping drive oncogenesis in 8-9% and 3% of non-small cell lung cancers (NSCLC) respectively. Their detection are essential for the management of patients since they confer sensitivity to specific targeted therapies with significant clinical benefit over conventional chemotherapy. Immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH) account for historical reference techniques however molecular-based technologies (RNA-based sequencing and RT-PCR) are emerging as alternative or complementary methods. Here, we evaluated the analytical performance of the fully-automated RT-PCR Idylla GeneFusion assay compared to reference methods using 35 fixed NSCLC samples. Idylla demonstrated overall agreement, sensitivity and specificity of 100% compared to RNASeq. Interestingly, it succeeded in retrieving 10 out of 11 samples with inconclusive results due to insufficient RNA quality for sequencing. Idylla showed an overall agreement, sensitivity and specificity of 90.32%, 91.67% and 89.47% compared to IHC/FISH respectively. Using commercial standards, the limit of detection of the Idylla system for the most frequent fusions and exon skipping ranges between 5 and 10 ng RNA input. These results support that the Idylla assay is a reliable and rapid option for the detection of these alterations, however a particular attention is needed for the interpretation of the expression imbalance.
Collapse
Affiliation(s)
- Pauline Gilson
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France.
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France.
| | - Celso Pouget
- Service d'Anatomocytopathologie, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Richard Belmonte
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Smahane Fadil
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Jessica Demange
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Marie Rouyer
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Julien Lacour
- Service d'Anatomocytopathologie, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Margaux Betz
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Julie Dardare
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Andréa Witz
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Jean-Louis Merlin
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Alexandre Harlé
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| |
Collapse
|
13
|
Wang C, Yuan X, Xue J. Targeted therapy for rare lung cancers: Status, challenges, and prospects. Mol Ther 2023; 31:1960-1978. [PMID: 37179456 PMCID: PMC10362419 DOI: 10.1016/j.ymthe.2023.05.007] [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: 03/02/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023] Open
Abstract
Lung cancer causes the most cancer-related deaths worldwide. In recent years, molecular and immunohistochemical techniques have rapidly developed, further inaugurating an era of personalized medicine for lung cancer. The rare subset of lung cancers accounts for approximately 10%, each displaying distinct clinical characteristics. Treatments for rare lung cancers are mainly based on evidence from common counterparts, which may lead to unsolid clinical benefits considering intertumoral heterogeneity. The increasing knowledge of molecular profiling of rare lung cancers has made targeting genetic alterations and immune checkpoints a powerful strategy. Additionally, cellular therapy has emerged as a promising way to target tumor cells. In this review, we first discuss the current status of targeted therapy and preclinical models for rare lung cancers, as well as provide mutational profiles by integrating the results of existing cohorts. Finally, we point out the challenges and future directions for developing targeted agents for rare lung cancer.
Collapse
Affiliation(s)
- Chunsen Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, the National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiang Yuan
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, the National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, the National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Van Herpe F, Van Cutsem E. The Role of cMET in Gastric Cancer—A Review of the Literature. Cancers (Basel) 2023; 15:cancers15071976. [PMID: 37046637 PMCID: PMC10093530 DOI: 10.3390/cancers15071976] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/10/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Gastric cancer (GC) is an important cause of cancer worldwide with over one million new cases yearly. The vast majority of cases present in stage IV disease, and it still bears a poor prognosis. However, since 2010, progress has been made with the introduction of targeted therapies against HER2 and with checkpoint inhibitors (PDL1). More agents interfering with other targets (FGFR2B, CLDN18.2) are being investigated. cMET is a less frequent molecular target that has been studied for gastric cancer. It is a proto-oncogene that leads to activation of the MAPK pathway and the PI3K pathway, which is responsible for activating the MTOR pathway. The prevalence of cMET is strongly debated as different techniques are being used to detect MET-driven tumors. Because of the difference in diagnostic assays, selecting patients who benefit from cMET inhibitors is difficult. In this review, we discuss the pathway of cMET, its clinical significance and the different diagnostic assays that are currently used, such as immunohistochemy (IHC), fluorescence in situ hybridization (FISH), the H-score and next-generation sequencing (NGS). Next, we discuss all the current data on cMET inhibitors in gastric cancer. Since the data on cMET inhibitors are very heterogenous, it is difficult to provide a general consensus on the outcome, as inclusion criteria differ between trials. Diagnosing cMET-driven gastric tumors is difficult, and potentially the only accurate determination of cMET overexpression/amplification may be next-generation sequencing (NGS).
Collapse
|
15
|
Kemper M, Krekeler C, Menck K, Lenz G, Evers G, Schulze AB, Bleckmann A. Liquid Biopsies in Lung Cancer. Cancers (Basel) 2023; 15:1430. [PMID: 36900221 PMCID: PMC10000706 DOI: 10.3390/cancers15051430] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
As lung cancer has the highest cancer-specific mortality rates worldwide, there is an urgent need for new therapeutic and diagnostic approaches to detect early-stage tumors and to monitor their response to the therapy. In addition to the well-established tissue biopsy analysis, liquid-biopsy-based assays may evolve as an important diagnostic tool. The analysis of circulating tumor DNA (ctDNA) is the most established method, followed by other methods such as the analysis of circulating tumor cells (CTCs), microRNAs (miRNAs), and extracellular vesicles (EVs). Both PCR- and NGS-based assays are used for the mutational assessment of lung cancer, including the most frequent driver mutations. However, ctDNA analysis might also play a role in monitoring the efficacy of immunotherapy and its recent accomplishments in the landscape of state-of-the-art lung cancer therapy. Despite the promising aspects of liquid-biopsy-based assays, there are some limitations regarding their sensitivity (risk of false-negative results) and specificity (interpretation of false-positive results). Hence, further studies are needed to evaluate the usefulness of liquid biopsies for lung cancer. Liquid-biopsy-based assays might be integrated into the diagnostic guidelines for lung cancer as a tool to complement conventional tissue sampling.
Collapse
Affiliation(s)
- Marcel Kemper
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| | - Carolin Krekeler
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| | - Kerstin Menck
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| | - Georg Lenz
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| | - Georg Evers
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| | - Arik Bernard Schulze
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| | - Annalen Bleckmann
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
| |
Collapse
|
16
|
Qin K, Hong L, Zhang J, Le X. MET Amplification as a Resistance Driver to TKI Therapies in Lung Cancer: Clinical Challenges and Opportunities. Cancers (Basel) 2023; 15:612. [PMID: 36765572 PMCID: PMC9913224 DOI: 10.3390/cancers15030612] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
Targeted therapy has emerged as an important pillar for the standard of care in oncogene-driven non-small cell lung cancer (NSCLC), which significantly improved outcomes of patients whose tumors harbor oncogenic driver mutations. However, tumors eventually develop resistance to targeted drugs, and mechanisms of resistance can be diverse. MET amplification has been proven to be a driver of resistance to tyrosine kinase inhibitor (TKI)-treated advanced NSCLC with its activation of EGFR, ALK, RET, and ROS-1 alterations. The combined therapy of MET-TKIs and EGFR-TKIs has shown outstanding clinical efficacy in EGFR-mutated NSCLC with secondary MET amplification-mediated resistance in a series of clinical trials. In this review, we aimed to clarify the underlying mechanisms of MET amplification-mediated resistance to tyrosine kinase inhibitors, discuss the ways and challenges in the detection and diagnosis of MET amplifications in patients with metastatic NSCLC, and summarize the recently published clinical data as well as ongoing trials of new combination strategies to overcome MET amplification-mediated TKI resistance.
Collapse
Affiliation(s)
- Kang Qin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lingzhi Hong
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
17
|
Oh SY, Lee YW, Lee EJ, Kim JH, Park Y, Heo SG, Yu MR, Hong MH, DaSilva J, Daly C, Cho BC, Lim SM, Yun MR. Preclinical Study of a Biparatopic METxMET Antibody-Drug Conjugate, REGN5093-M114, Overcomes MET-driven Acquired Resistance to EGFR TKIs in EGFR-mutant NSCLC. Clin Cancer Res 2023; 29:221-232. [PMID: 36269795 DOI: 10.1158/1078-0432.ccr-22-2180] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/08/2022] [Accepted: 10/19/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE MET amplification is a frequent mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI) in patients with EGFR-mutated non-small cell lung cancer (NSCLC), and combined treatment with EGFR TKIs and MET TKIs has been explored as a strategy to overcome resistance. However, durable response is invariably limited by the emergence of acquired resistance. Here, we investigated the preclinical activity of REGN5093-M114, a novel antibody-drug conjugate targeting MET in MET-driven patient-derived models. EXPERIMENTAL DESIGN Patient-derived organoids, patient-derived cells, or ATCC cell lines were used to investigate the in vitro/in vivo activity of REGN5093-M114. RESULTS REGN5093-M114 exhibited significant antitumor efficacy compared with MET TKI or unconjugated METxMET biparatopic antibody (REGN5093). Regardless of MET gene copy number, MET-overexpressed TKI-naïve EGFR-mutant NSCLC cells responded to REGN5093-M114 treatment. Cell surface MET expression had the most predictive power in determining the efficacy of REGN5093-M114. REGN5093-M114 potently reduced tumor growth of EGFR-mutant NSCLC with PTEN loss or MET Y1230C mutation after progression on prior osimertinib and savolitinib treatment. CONCLUSIONS Altogether, REGN5093-M114 is a promising candidate to overcome the challenges facing functional MET pathway blockade.
Collapse
Affiliation(s)
- Seung Yeon Oh
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Department of Biomedical Science institute, Graduated School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - You Won Lee
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Eun Ji Lee
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Department of Biomedical Science institute, Graduated School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Jae Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - YoungJoon Park
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Seong Gu Heo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Mi Ra Yu
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - John DaSilva
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Mi Ran Yun
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of South Korea
| |
Collapse
|
18
|
Longitudinal Plasma Proteomics-Derived Biomarkers Predict Response to MET Inhibitors for MET-Dysregulated NSCLC. Cancers (Basel) 2023; 15:cancers15010302. [PMID: 36612298 PMCID: PMC9818927 DOI: 10.3390/cancers15010302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
MET inhibitors have shown promising efficacy for MET-dysregulated non-small cell lung cancer (NSCLC). However, quite a few patients cannot benefit from it due to the lack of powerful biomarkers. This study aims to explore the potential role of plasma proteomics-derived biomarkers for patients treated with MET inhibitors using mass spectrometry. We analyzed the plasma proteomics from patients with MET dysregulation (including MET amplification and MET overexpression) treated with MET inhibitors. Thirty-three MET-dysregulated NSCLC patients with longitudinal 89 plasma samples were included. We classified patients into the PD group and non-PD group based on clinical response. The baseline proteomic profiles of patients in the PD group were distinct from those in the non-PD group. Through protein screening, we found that a four-protein signature (MYH9, GNB1, ALOX12B, HSD17B4) could predict the efficacy of patients treated with MET inhibitors, with an area under the curve (AUC) of 0.93, better than conventional fluorescence in situ hybridization (FISH) or immunohistochemistry (IHC) tests. In addition, combining the four-protein signature with FISH or IHC test could also reach higher predictive performance. Further, the combined signature could predict progression-free survival for MET-dysregulated NSCLC (p < 0.001). We also validated the performance of the four-protein signature in another cohort of plasma using an enzyme-linked immunosorbent assay. In conclusion, the four plasma protein signature (MYH9, GNB1, ALOX12B, and HSD17B4 proteins) might play a substitutable or complementary role to conventional MET FISH or IHC tests. This exploration will help select patients who may benefit from MET inhibitors.
Collapse
|
19
|
Wu Y, Yan Z, Pan J, Chang X, Huang B, Luo D, Meng R, Shi H, Fan J, Nie X. Partial response to pralsetinib in an advanced pulmonary sarcomatoid carcinoma patient harboring a KIF5B-RET rearrangement: a case report. World J Surg Oncol 2022; 20:386. [PMID: 36471407 PMCID: PMC9724403 DOI: 10.1186/s12957-022-02848-z] [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: 06/29/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pulmonary sarcomatoid carcinoma (PSC) is a rare and unconventional non-small-cell lung cancer (NSCLC) that appears to be aggressive, with a poor prognosis and response to conventional treatment. Approximately 30% of PSCs have potentially targetable genomic alterations, but few studies have involved RET gene fusions, and corresponding targeted therapies are lacking. CASE PRESENTATION In this report, we describe a patient with PSC harboring a KIF5B-RET gene fusion who was initially diagnosed with stage IVb lung cancer. Due to the poor performance status, the patient was unable to tolerate any radiotherapy or chemotherapy. Based on the next-generation sequencing (NGS) result of RET gene fusion, the patient was treated with pralsetinib. Two months after the treatment, the patient achieved a partial response. CONCLUSIONS Our case indicates that RET is one of the main driver oncogenes of PSC and provides useful information for precise RET inhibitor administration in the future. Thus, the use of comprehensive genomic profiling may provide important treatment options for PSC.
Collapse
Affiliation(s)
- Ying Wu
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Zhecheng Yan
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Juan Pan
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Xiaona Chang
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Bo Huang
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Danju Luo
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Rui Meng
- grid.33199.310000 0004 0368 7223Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heshui Shi
- grid.33199.310000 0004 0368 7223Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Jun Fan
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Xiu Nie
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| |
Collapse
|
20
|
MET inhibitors for MET amplification-What's the most appropriate detection method and cut-off value? Lung Cancer 2022; 174:172-173. [PMID: 35985889 DOI: 10.1016/j.lungcan.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/30/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022]
|
21
|
Moiseenko F, Bogdanov A, Egorenkov V, Volkov N, Moiseyenko V. Management and Treatment of Non-small Cell Lung Cancer with MET Alteration and Mechanisms of Resistance. Curr Treat Options Oncol 2022; 23:1664-1698. [PMID: 36269457 DOI: 10.1007/s11864-022-01019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT MET-driven tumors are a heterogenous group of non-small cell lung cancers (NSCLC) with activating mutations. Pathologic activation of MET can be achieved with increased number of gene copies overexpression, or decreased protein degradation through several mechanisms, including mutations, amplifications, or fusions. Besides its role as primary driver, MET activation might also mediate resistance to kinase inhibitors in NSCLC with various other actionable alterations. While checkpoint inhibitors have modest efficacy in MET-driven tumors, several approaches of targeted blockade are available. Among them the most promising are small tyrosine kinase inhibitors, antibody-drug conjugates, and bispecific antibodies. Unfortunately, resistance is virtually inevitable. Resistance to small kinase inhibitors might be mediated by kinase domain mutations or activation of shunting cascades. Various resistance mechanisms might be present in one patient, making it overcoming an unresolved problem.
Collapse
Affiliation(s)
- Fedor Moiseenko
- Saint-Petersburg City Cancer Center, Leningradskay 68a, Lit.A, Pesochny, St. Petersburg, 197758, Russia. .,N.N. Petrov National Medical Research Center of Oncology, Ministry of Public Health of the Russian Federation, 68, Leningradskaya st., Pesochny, St. Petersburg, 197758, Russia. .,State Budget Institution of Higher Education "North-Western State Medical University named after I.I Mechnikov" under the Ministry of Public Health of the Russian Federation, 41, Kirochnaya str, Saint Petersburg, 191015, Russia.
| | - Alexey Bogdanov
- Saint-Petersburg City Cancer Center, Leningradskay 68a, Lit.A, Pesochny, St. Petersburg, 197758, Russia
| | - Vitaliy Egorenkov
- Saint-Petersburg City Cancer Center, Leningradskay 68a, Lit.A, Pesochny, St. Petersburg, 197758, Russia
| | - Nikita Volkov
- Saint-Petersburg City Cancer Center, Leningradskay 68a, Lit.A, Pesochny, St. Petersburg, 197758, Russia
| | - Vladimir Moiseyenko
- Saint-Petersburg City Cancer Center, Leningradskay 68a, Lit.A, Pesochny, St. Petersburg, 197758, Russia
| |
Collapse
|
22
|
Michaels E, Bestvina CM. Meeting an un-MET need: Targeting MET in non-small cell lung cancer. Front Oncol 2022; 12:1004198. [PMID: 36338701 PMCID: PMC9634070 DOI: 10.3389/fonc.2022.1004198] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
The MET pathway can be activated by MET exon 14 skipping mutations, gene amplification, or overexpression. Mutations within this pathway carry a poor prognosis for patients with non-small cell lung cancer (NSCLC). MET exon 14 skipping mutations occur in 3-4% of patients with NSCLC, while MET amplifications are found in 1-6% of patients. The most effective method for detection of MET amplification is fluorescent in situ hybridization (FISH) and of MET exon 14 skipping mutations is RNA-based next generation sequencing (NGS). Immunohistochemistry (IHC) is an alternative method of diagnosis but is not as reliable. Early studies of MET tyrosine kinase inhibitors (TKIs) demonstrated limited clinical benefit. However, newer selective MET TKIs, such as capmatinib and tepotinib, have improved efficacy. Both drugs have an acceptable safety profile with the most common treatment-related adverse event being peripheral edema. One of the most frequent resistance mechanisms to EGFR inhibition with osimertinib is MET amplification. There is interest in combining EGFR inhibition plus MET inhibition in an attempt to target this resistance mechanism. Additional ways of targeting MET alterations are currently under investigation, including the bi-specific antibody amivantamab. Additional research is needed to further understand resistance mechanisms to MET inhibition. There is limited research into the efficacy of immune checkpoint inhibition for MET-altered NSCLC, though some data suggests decreased efficacy compared with wild-type patients and increased toxicity associated with the combination of immunotherapy and MET TKIs. Future directions for research will include combination clinical trials and understanding rational combinations for MET alterations.
Collapse
Affiliation(s)
- Elena Michaels
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Christine M. Bestvina
- Department of Medicine, University of Chicago Comprehensive Cancer Center, Chicago, IL, United States
| |
Collapse
|
23
|
Remon J, Hendriks LE, Mountzios G, García-Campelo R, Saw SP, Uprety D, Recondo G, Villacampa G, Reck M. MET alterations in NSCLC—Current Perspectives and Future Challenges. J Thorac Oncol 2022; 18:419-435. [PMID: 36441095 DOI: 10.1016/j.jtho.2022.10.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 10/19/2022] [Indexed: 11/24/2022]
Abstract
Targeted therapies have revolutionized the treatment and improved the outcome for oncogene-driven NSCLC and an increasing number of oncogenic driver therapies have become available. For MET-dysregulated NSCLC (especially MET exon 14 skipping mutations and MET-amplifications, which is one of the most common bypass mechanisms of resistance in oncogene-addicted NSCLC), several anti-MET-targeted therapies have been approved recently (MET exon 14 skipping mutation) and multiple others are in development. In this narrative review, we summarize the role of MET as an oncogenic driver in NSCLC, discuss the different testing methods for exon 14 skipping mutations, gene amplification, and protein overexpression, and review the existing data and ongoing clinical trials regarding targeted therapies in MET-altered NSCLC. As immunotherapy with or without chemotherapy has become the standard of care for advanced NSCLC, immunotherapy data for MET-dysregulated NSCLC are put into perspective. Finally, we discuss future challenges in this rapidly evolving landscape.
Collapse
|
24
|
Zhao C, Gao S, Xue Q, Tan F, Gao Y, Mao Y, Wang D, Zhao J, Yang D, Hong Q, Mu J. Clinical characteristics and prognostic factors of pulmonary sarcomatoid carcinoma. J Thorac Dis 2022; 14:3773-3781. [PMID: 36389311 PMCID: PMC9641323 DOI: 10.21037/jtd-22-393] [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: 03/24/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Pulmonary sarcomatoid carcinoma (PSC) is a kind of rare lung cancer. We aim to analyze the clinical characteristics and prognostic factors of patients with PSC. METHODS From January 1, 2006 to December 31, 2015, 119 patients in the Cancer Hospital Chinese Academy of Medical Sciences were diagnosed with PSC, and they received treatment. We retrospectively collected information on gender, age, body mass index (BMI), symptoms, family history, smoking history, tumor size, tumor location, tumor diameter, tumor-node-metastasis (TNM), pathological type, and other factors to analyze the relationship between these factors and 1-, 3-, 5-year, and overall survival (OS). RESULTS Male patients who had a smoking history (n=76) comprised the main group of PSC. Median patient age was 60.67±10.50 years (range, 26-89 years). The majority of these patients (n=82) presented with respiratory symptoms. The median survival of patients who died of PSC was 11.87 months (6.38-21.48 months). The 1-, 3-, and 5-year survival rates were 61.3%, 34.5%, and 31.9%, respectively. Patients with a lower T stage and without lymph node metastasis and distant metastasis had a better OS (P<0.05). Other clinical characteristics and the difference in treatments did not influence the prognosis significantly (P>0.05). CONCLUSIONS PSC is a rare malignant neoplasm of the lung with poor prognosis. Surgery is a major therapeutic method for this disease entity. TNM-stage is the main factors affecting prognosis.
Collapse
Affiliation(s)
- Chenguang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yousheng Mao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dali Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ding Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Hong
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juwei Mu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Gong C, Xiong H, Qin K, Wang J, Cheng Y, Zhao J, Zhang J. MET alterations in advanced pulmonary sarcomatoid carcinoma. Front Oncol 2022; 12:1017026. [PMID: 36212500 PMCID: PMC9539670 DOI: 10.3389/fonc.2022.1017026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/09/2022] [Indexed: 11/23/2022] Open
Abstract
Pulmonary sarcomatoid carcinoma (PSC) is a rare subset of NSCLC that accounts for about 0.5-1% of all primary lung carcinoma, and its malignant biological behavior is more aggressive than other pathological types of lung cancer. Recent studies have reported a variety of gene mutations associated with the occurrence, development and treatment of PSC, especially the mesenchymal-epithelial transition (MET) proto-oncogene alterations, including the exon 14 (METex14) skipping mutations as well as the amplification and overexpression of MET gene, which are associated with molecularly targeted therapy for PSC. METex14 skipping mutation is the most common and well-studied mutation type, occurring in about 22-31.8% of PSC patients, while the prevalence of MET amplification is reported as 4.8-13.6% and MET ovexpression is about 20.2%. Molecular pathology tests, including IHC and NGS, are valuable in determining the prognosis of patients with PSC and helping to determine the treatment. The existing clinical data have confirmed the efficacy of MET-TKI in PSC patients with MET alteration, among which the clinical study of Savolitinib has enrolled the largest proportion of PSC patients and achieved relatively good efficacy, but more clinical researches are still needed. The multi-disciplinary team may maximize the optimal treatment options for patients with the advanced PSC.
Collapse
|
26
|
Wang N, Zhu Y, Wu Y, Huang B, Wu J, Zhang R, Fan J, Nie X. MET overexpression in EGFR L858R mutant treatment-naïve advanced lung adenocarcinoma correlated with poor prognosis: a real-world retrospective study. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04225-5. [PMID: 35904603 DOI: 10.1007/s00432-022-04225-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/18/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND Mesenchymal epithelial transition (MET) overexpression has been reported in approximately 50-60% of epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancers. However, the prognostic significance of MET overexpression has not been established in advanced lung adenocarcinoma (ADC) patients with EGFR-sensitive mutations. METHODS A retrospective study was performed on a total of 406 treatment-naïve advanced ADC patients with EGFR mutation detection and MET expression information. EGFR mutations were detected by next-generation sequencing or amplification refractory mutation system-polymerase chain reaction. Immuno-histochemistry staining of MET expression was evaluated by H-score and overexpression was defined as an H-score ≥ 200. Overall survival (OS) and progression-free survival (PFS) were analyzed according to MET expression. RESULTS Among the 406 patients, 208 patients had EGFR mutations, including 102 exon 19_del mutations, 94 L858R mutations and 12 other types of mutations. Of 110 patients with concomitant EGFR mutations and MET overexpression, 61 (59.8%) patients had 19_del mutations, 44 (46.8%) patients had L858R mutations and five (41.7%) patients had others. Patients with MET overexpression had a markedly shorter PFS and OS than patients with MET H-score < 200 in the EGFR L858R mutation subgroup (median PFS: 12 versus 26 months, p = 0.001; median OS: 24 versus 32 months, p = 0.038), whereas no significant difference was observed in 19_del mutation subgroup. Multivariate Cox analysis showed that MET overexpression was an independent poor prognostic factor for PFS and OS in patients with the L858R mutations (HR = 3.064, 95% CI 1.705-5.507, p < 0.001; HR = 2.043, 95% CI 1.000-4.172; p = 0.049), rather than 19_del. CONCLUSIONS MET overexpression is a poor prognostic factor for advanced ADC patients with the EGFR L858R mutation.
Collapse
Affiliation(s)
- Na Wang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Yili Zhu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Ying Wu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Bo Huang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Junhua Wu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Ruiguang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jun Fan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
| | - Xiu Nie
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
27
|
MET Expression Level in Lung Adenocarcinoma Loosely Correlates with MET Copy Number Gain/Amplification and Is a Poor Predictor of Patient Outcome. Cancers (Basel) 2022; 14:cancers14102433. [PMID: 35626038 PMCID: PMC9139916 DOI: 10.3390/cancers14102433] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/02/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary MET is a proto-oncogene and plays an important role on tumor cell survival, proliferation, metastasis, and drug resistance. Patient with MET amplification has shown an inferior outcome comparing to patients without MET amplification. Fluorescence in situ hybridization (FISH) is often used to detect MET amplification, and immunohistochemistry (IHC) is often used to assess MET expression level. Though some institutions provide both tests, IHC is more readily available in most pathology laboratories and is cheaper than FISH. This study evaluated the correlation of MET expression level with MET copy number gain/amplification, and the MET overexpression with patient’s outcome. By studying 446 patients with lung adenocarcinoma, we found that the concordance of MET expression and MET copy number gain/amplification was low; high-level of MET expression was associated with inferior outcome, but it was not an independent poor prognostic factor. These findings indicate that IHC for MET expression can’t substitute FISH analysis for MET amplification. Abstract MET amplification has been associated with shorter survival in cancer patients, however, the potential correlation of MET overexpression with either MET amplification or patient outcome is controversial. The aim of this study was to address these questions by correlating MET expression level with MET copy number and patient outcome in a cohort of 446 patients who had a lung adenocarcinoma: 88 with MET amplification, 118 with polysomy 7, and 240 with negative results by fluorescence in situ hybridization. MET expression assessed by immunohistochemistry was semi-quantified by expression level: absent (0+), weak (1+), moderate (2+) and strong (3+); or by H-score: 0–99, 100–199, and ≥200. MET expression level or H-score was positively but weakly correlated with MET copy number or MET/CEP7 ratio. Strong expression of MET (3+ or H-score ≥ 200) was associated with a shorter overall survival, but it was not an independent hazard for survival by multivariant analysis. We conclude that MET expression is loosely correlated with MET copy number gain/amplification. Strong expression of MET does not independently predict patient outcome.
Collapse
|
28
|
Cheema PK, Banerji SO, Blais N, Chu QSC, Desmeules P, Juergens RA, Leighl NB, Sheffield BS, Wheatley-Price PF, Melosky BL. Canadian Consensus Recommendations on the Management of MET-Altered NSCLC. Curr Oncol 2021; 28:4552-4576. [PMID: 34898564 PMCID: PMC8628757 DOI: 10.3390/curroncol28060386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/29/2022] Open
Abstract
In Canada, the therapeutic management of patients with advanced non-small cell lung cancer (NSCLC) with rare actionable mutations differs between provinces, territories, and individual centres based on access to molecular testing and funded treatments. These variations, together with the emergence of several novel mesenchymal-epithelial transition (MET) factor-targeted therapies for the treatment of NSCLC, warrant the development of evidence-based consensus recommendations for the use of these agents. A Canadian expert panel was convened to define key clinical questions, review evidence, discuss practice recommendations and reach consensus on the treatment of advanced MET-altered NSCLC. Questions addressed by the panel include: 1. How should the patients most likely to benefit from MET-targeted therapies be identified? 2. What are the preferred first-line and subsequent therapies for patients with MET exon 14 skipping mutations? 3. What are the preferred first-line and subsequent therapies for advanced NSCLC patients with de novo MET amplification? 4. What is the preferred therapy for patients with advanced epidermal growth factor receptor (EGFR)-mutated NSCLC with acquired MET amplification progressing on EGFR inhibitors? 5. What are the potential strategies for overcoming resistance to MET inhibitors? Answers to these questions, along with the consensus recommendations herein, will help streamline the management of MET-altered NSCLC in routine practice, assist clinicians in therapeutic decision-making, and help ensure optimal outcomes for NSCLC patients with MET alterations.
Collapse
Affiliation(s)
- Parneet K. Cheema
- Medical Oncology/Hematology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shantanu O. Banerji
- CancerCare Manitoba Research Institute, Department of Medical Oncology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Normand Blais
- Department of Medicine, Centre Hospitalier de l’Université de Montréal, University of Montreal, Montreal, QC H2X 3E4, Canada;
| | - Quincy S.-C. Chu
- Cross Cancer Institute, Alberta Health Services, Edmonton, AB T6G 1Z2, Canada;
| | - Patrice Desmeules
- Service d’Anatomopathologie et de Cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC G1V 0A6, Canada;
| | - Rosalyn A. Juergens
- Department of Medical Oncology, Juravinski Cancer Centre, McMaster University, Hamilton, ON L8V 5C2, Canada;
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University Health Network, Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Brandon S. Sheffield
- Department of Laboratory Medicine, William Osler Health System, Brampton, ON L6R 3J7, Canada;
| | - Paul F. Wheatley-Price
- Department of Medicine, The Ottawa Hospital Research Institute, The Ottawa Hospital, University of Ottawa, Ottawa, ON K1H 8L6, Canada;
| | - Barbara L. Melosky
- Department of Medical Oncology, BC Cancer-Vancouver Centre, Vancouver, BC V5Z 4E6, Canada;
| |
Collapse
|
29
|
Chen M, Yang Q, Xu Z, Luo B, Li F, Yu Y, Sun J. Survival Analysis and Prediction Model for Pulmonary Sarcomatoid Carcinoma Based on SEER Database. Front Oncol 2021; 11:630885. [PMID: 34136380 PMCID: PMC8201495 DOI: 10.3389/fonc.2021.630885] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
Objective This study aimed to investigate the incidence of the pulmonary sarcomatoid carcinoma (PSC), to compare the clinical characteristics and overall survival (OS) of patients with PSC and those with other non-small-cell lung cancer (oNSCLC), so as to analyze the factors affecting the OS of patients with PSC and construct a nomogram prediction model. Methods Data of patients with PSC and those with oNSCLC diagnosed between 2004 and 2015 from the Surveillance, Epidemiology, and End Results database were collected. The age-adjusted incidence of PSC was calculated. The characteristics of patients with PSC and those with oNSCLC were compared, then the patients were matched 1:2 for further survival analysis. Patients with PSC were randomly divided into training set and testing set with a ratio of 7:3. The Cox proportional hazards model was used to identify the covariates associated with the OS. Significant covariates were used to construct the nomogram, and the C-index was calculated to measure the discrimination ability. The accuracy of the nomogram was compared with the tumor–node–metastasis (TNM) clinical stage, and the corresponding area under the curve was achieved. Results A total of 1049 patients with PSC were enrolled, the incidence of PSC was slowly decreased from 0.120/100,000 in 2004 to 0.092/100,000 in 2015. Before PSM, 793 PSC patients and 191356 oNSCLC patients were identified, the proportion of male, younger patients (<65 years), grade IV, TNM clinical stage IV was higher in the PSC. The patients with PSC had significantly poorer OS compared with those with oNSCLC. After PSM, PSC still had an extremely inferior prognosis. Age, sex, TNM clinical stage, chemotherapy, radiotherapy, and surgery were independent factors for OS. Next, a nomogram was established based on these factors, and the C-indexs were 0.775 and 0.790 for the training and testing set, respectively. Moreover, the nomogram model indicated a more comprehensive and accurate prediction than the TNM clinical stage. Conclusions The incidence of PSC was slowly decreased. PSC had a significantly poor prognosis compared with oNSCLC. The nomogram constructed in this study accurately predicted the prognosis of PSC, performed better than the TNM clinical stage.
Collapse
Affiliation(s)
- Mingjing Chen
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China.,Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiao Yang
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China.,Department of Ultrasound, The 941st Hospital of the PLA Joint Logistic Support Force, Xining, China
| | - Zihan Xu
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China.,Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Bangyu Luo
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Feng Li
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yongxin Yu
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
30
|
Subramanian J, Tawfik O. Detection of MET exon 14 skipping mutations in non-small cell lung cancer: overview and community perspective. Expert Rev Anticancer Ther 2021; 21:877-886. [PMID: 33957836 DOI: 10.1080/14737140.2021.1924683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Non-small cell lung cancer (NSCLC), which accounts for the majority of lung cancer diagnoses in the United States, has many known driver mutations, including MET exon 14 skipping mutation (METex14). The detection of oncogenic driver mutations in NSCLC and the development of drugs to target these alterations, including METex14, has created the need for accurate and reliable testing, of which next-generation sequencing (NGS) is the gold standard. However, detection of METex14 in patients with NSCLC can be challenging due to the complex biology of METex14 and the abilities of different NGS platforms to detect METex14.Areas covered: This review provides an overview of METex14 biology, discusses the optimal platforms for the detection of METex14 in NSCLC, and provides an overview of the use of NGS in the community setting.Expert opinion: Broad molecular testing is crucial for identifying actionable oncogenic drivers in NSCLC. METex14 is a complex oncogenic driver mutation requiring carefully optimized platforms for proper detection. To identify patients eligible for targeted therapies - including therapies targeting novel oncogenic drivers, such as MET inhibitors - community oncologists need to be aware of both the use of NGS platforms and the differences in their capabilities to detect certain oncogenic drivers.
Collapse
Affiliation(s)
- Janakiraman Subramanian
- Department of Medicine, University of Missouri-Kansas City, School of Medicine, Kansas City, Missouri, USA.,Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri, USA
| | - Ossama Tawfik
- Department of Pathology, Saint Luke's Health System of Kansas City, MAWD Pathology Group, Lenexa, Kansas, USA
| |
Collapse
|
31
|
Socinski MA, Pennell NA, Davies KD. MET Exon 14 Skipping Mutations in Non-Small-Cell Lung Cancer: An Overview of Biology, Clinical Outcomes, and Testing Considerations. JCO Precis Oncol 2021; 5:PO.20.00516. [PMID: 34036238 PMCID: PMC8140815 DOI: 10.1200/po.20.00516] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Nathan A. Pennell
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Kurtis D. Davies
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
32
|
Tan AC, Loh TJ, Kwang XL, Tan GS, Lim KH, Tan DSW. Novel Therapies for Metastatic Non-Small Cell Lung Cancer with MET Exon 14 Alterations: A Spotlight on Capmatinib. LUNG CANCER-TARGETS AND THERAPY 2021; 12:11-20. [PMID: 33776501 PMCID: PMC7987308 DOI: 10.2147/lctt.s263610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
MET exon 14 (METex14) alterations are now an established therapeutically tractable target in non-small cell lung cancer (NSCLC). Recently reported trials of several MET tyrosine kinase inhibitors (TKI) in this patient population have demonstrated promising efficacy data in both the treatment naïve and pre-treated settings and have led to regulatory approvals. This review will focus on practical diagnostic considerations for METex14 alterations, the trial evidence for capmatinib in this molecular subset including dosing and toxicity management, and the future therapeutic landscape of METex14 altered NSCLC.
Collapse
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Tracy J Loh
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Xue Lin Kwang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Gek San Tan
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| |
Collapse
|
33
|
Schmidt U, Heller G, Timelthaler G, Heffeter P, Somodi Z, Schweifer N, Sibilia M, Berger W, Csiszar A. The FAM3C locus that encodes interleukin-like EMT inducer (ILEI) is frequently co-amplified in MET-amplified cancers and contributes to invasiveness. J Exp Clin Cancer Res 2021; 40:69. [PMID: 33596971 PMCID: PMC7890988 DOI: 10.1186/s13046-021-01862-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gene amplification of MET, which encodes for the receptor tyrosine kinase c-MET, occurs in a variety of human cancers. High c-MET levels often correlate with poor cancer prognosis. Interleukin-like EMT inducer (ILEI) is also overexpressed in many cancers and is associated with metastasis and poor survival. The gene for ILEI, FAM3C, is located close to MET on chromosome 7q31 in an amplification "hotspot", but it is unclear whether FAMC3 amplification contributes to elevated ILEI expression in cancer. In this study we have investigated FAMC3 copy number gain in different cancers and its potential connection to MET amplifications. METHODS FAMC3 and MET copy numbers were investigated in various cancer samples and 200 cancer cell lines. Copy numbers of the two genes were correlated with mRNA levels, with relapse-free survival in lung cancer patient samples as well as with clinicopathological parameters in primary samples from 49 advanced stage colorectal cancer patients. ILEI knock-down and c-MET inhibition effects on proliferation and invasiveness of five cancer cell lines and growth of xenograft tumors in mice were then investigated. RESULTS FAMC3 was amplified in strict association with MET amplification in several human cancers and cancer cell lines. Increased FAM3C and MET copy numbers were tightly linked and correlated with increased gene expression and poor survival in human lung cancer and with extramural invasion in colorectal carcinoma. Stable ILEI shRNA knock-down did not influence proliferation or sensitivity towards c-MET-inhibitor induced proliferation arrest in cancer cells, but impaired both c-MET-independent and -dependent cancer cell invasion. c-MET inhibition reduced ILEI secretion, and shRNA mediated ILEI knock-down prevented c-MET-signaling induced elevated expression and secretion of matrix metalloproteinase (MMP)-2 and MMP-9. Combination of ILEI knock-down and c-MET-inhibition significantly reduced the invasive outgrowth of NCI-H441 and NCI-H1993 lung tumor xenografts by inhibiting proliferation, MMP expression and E-cadherin membrane localization. CONCLUSIONS These novel findings suggest MET amplifications are often in reality MET-FAM3C co-amplifications with tight functional cooperation. Therefore, the clinical relevance of this frequent cancer amplification hotspot, so far dedicated purely to c-MET function, should be re-evaluated to include ILEI as a target in the therapy of c-MET-amplified human carcinomas.
Collapse
Affiliation(s)
- Ulrike Schmidt
- Research Institute of Molecular Pathology, Dr. Bohr-Gasse 3, A-1030 Vienna, Austria
| | - Gerwin Heller
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Gerald Timelthaler
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Zsolt Somodi
- Department of Oncology, Bacs-Kiskun County Teaching Hospital, Kecskemet, Hungary
- Present Address: Parexel International, 2 Federal St, Billerica, MA USA
| | | | - Maria Sibilia
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Agnes Csiszar
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| |
Collapse
|
34
|
König D, Savic Prince S, Rothschild SI. Targeted Therapy in Advanced and Metastatic Non-Small Cell Lung Cancer. An Update on Treatment of the Most Important Actionable Oncogenic Driver Alterations. Cancers (Basel) 2021; 13:804. [PMID: 33671873 PMCID: PMC7918961 DOI: 10.3390/cancers13040804] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
Due to groundbreaking developments and continuous progress, the treatment of advanced and metastatic non-small cell lung cancer (NSCLC) has become an exciting, but increasingly challenging task. This applies, in particular, to the subgroup of NSCLC with oncogenic driver alterations. While the treatment of epidermal growth factor receptor (EGFR)-mutated and anaplastic lymphoma kinase (ALK)-rearranged NSCLC with various tyrosine kinase inhibitors (TKIs) is well-established, new targets have been identified in the last few years and new TKIs introduced in clinical practice. Even for KRAS mutations, considered for a long time as an "un-targetable" alteration, promising new drugs are emerging. The detection and in-depth molecular analysis of resistance mechanisms has further fueled the development of new therapeutic strategies. The objective of this review is to give a comprehensive overview on the current landscape of targetable oncogenic alterations in NSCLC.
Collapse
Affiliation(s)
- David König
- Department of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland;
- Comprehensive Cancer Center, University Hospital Basel, 4031 Basel, Switzerland;
| | - Spasenija Savic Prince
- Comprehensive Cancer Center, University Hospital Basel, 4031 Basel, Switzerland;
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland
| | - Sacha I. Rothschild
- Department of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland;
- Comprehensive Cancer Center, University Hospital Basel, 4031 Basel, Switzerland;
| |
Collapse
|
35
|
Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol 2021; 160:103234. [PMID: 33497758 DOI: 10.1016/j.critrevonc.2021.103234] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (including EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
36
|
Sui JSY, Finn SP, Gray SG. Detection of MET Exon 14 Skipping Alterations in Lung Cancer Clinical Samples Using a PCR-Based Approach. Methods Mol Biol 2021; 2279:145-155. [PMID: 33683691 DOI: 10.1007/978-1-0716-1278-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The receptor tyrosine kinase (RTK) c-MET plays important roles in cancer, yet despite being frequently overexpressed, clinical responses to targeting this receptor have been limited in the clinical setting. A singular significant challenge has been the accurate identification of biomarkers for the selection of responsive patients. However, recently mutations which result in the loss of exon 14 (called METex14 skipping) have emerged as novel biomarkers in non-small cell lung carcinomas (NSCLC) to predict for responsiveness to targeted therapy with c-MET inhibitors. Currently, the diverse genomic alterations responsible for METex14 skipping pose a challenge for routine clinical diagnostic testing. Next generation sequencing (NGS) is the current gold standard for identifying the diverse mutations associated with METex14, but the cost for such a procedure remains to some degree prohibitive as often NGS is requested on a case-by-case basis, and many hospitals may not even have the capacity or resources to conduct NGS.However, PCR-based approaches to detect METex14 have been developed which can be conducted in most routine hospital laboratories and may therefore allow a cost-effective approach to pre-screen patients that may respond to c-MET inhibitors prior to conducting NGS, or until all patients will have NGS conducted as routine practise. In this chapter, we describe one such PCR-based approach for screening samples for the detection of METex14 in NSCLC.
Collapse
Affiliation(s)
- Jane S Y Sui
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Stephen P Finn
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
- Department of Histopathology, Cancer Molecular Diagnostics, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin, Ireland
| | - Steven G Gray
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland.
- School of Biological Sciences, Dublin Institute of Technology, Dublin, Ireland.
| |
Collapse
|
37
|
Yin W, Cheng J, Tang Z, Toruner G, Hu S, Guo M, Robinson M, Medeiros LJ, Tang G. MET Amplification (MET/CEP7 Ratio ≥ 1.8) Is an Independent Poor Prognostic Marker in Patients With Treatment-naive Non-Small-cell Lung Cancer. Clin Lung Cancer 2020; 22:e512-e518. [PMID: 33288441 DOI: 10.1016/j.cllc.2020.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/26/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
INTRODUCTION The MET pathway is a promising target in patients with non-small-cell lung cancer (NSCLC). Fluorescence in situ hybridization analysis has become a standard method to detect MET amplification. However, no consensus has been reached regarding the definition of MET amplification. We aimed to find clinically meaningful cutoffs for MET amplification that could be used as a prognostic marker and/or indication for MET inhibitor therapy. PATIENTS AND METHODS We reviewed the fluorescence in situ hybridization results of MET/CEP7 (centromere of chromosome 7) for 2260 patients with treatment-naive NSCLC from 2014 to 2019. Clinical and pathologic data were collected from the medical records. Log-rank tests and Cox proportional hazard models were used to estimate the overall survival (OS) among patients with different MET/CEP7 ratios and/or MET copy numbers. RESULTS Of the 2260 patients, 130 (5.8%) had had a MET/CEP7 ratio of ≥ 1.8 and 13 (0.6%) had had a ratio of ≥ 5.0. Of these 130 patients with a MET/CEP7 ratio of ≥ 1.8, 123 (95%) also had a MET copy number of ≥ 5. In general, a higher MET copy number and higher MET/CEP7 ratio were associated with advanced tumor stage. The OS was significantly shorter when the MET copy number was ≥ 10 and/or when the MET/CEP7 ratio was ≥ 1.8. A MET/CEP7 ratio of ≥ 1.8 remained a significant hazard to OS on multivariate analysis (hazard ratio, 1.63; P = .019). CONCLUSIONS Patients with a MET copy number of ≥ 10 and/or MET/CEP7 ratio of ≥ 1.8 showed significantly poorer survival, and a MET/CEP7 ratio of ≥ 1.8 was an independent poor prognostic factor.
Collapse
Affiliation(s)
- Wei Yin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Joanne Cheng
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gokce Toruner
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shimin Hu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ming Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Melissa Robinson
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
38
|
Pharmaceutical strategies in the emerging era of antibody-based biotherapeutics for the treatment of cancers overexpressing MET receptor tyrosine kinase. Drug Discov Today 2020; 26:106-121. [PMID: 33171292 DOI: 10.1016/j.drudis.2020.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/23/2020] [Accepted: 11/03/2020] [Indexed: 12/26/2022]
Abstract
Pharmaceutical innovation in the development of novel antibody-based biotherapeutics with increased therapeutic indexes makes MET-targeted cancer therapy a clinical reality.
Collapse
|
39
|
Ding Y, Shao Y, Na C, Yin JC, Hua H, Tao R, Jiang Y, Hu R, He X, Miao C, Zhu D, Zhang Z. Genetic characterisation of sarcomatoid carcinomas reveals multiple novel actionable mutations and identifies KRAS mutation as a biomarker of poor prognosis. J Med Genet 2020; 59:10-17. [PMID: 33115932 DOI: 10.1136/jmedgenet-2020-107083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Sarcomatoid component occurs in various epithelial malignancies and is associated with an aggressive disease course and poor clinical outcome. As it is largely rare, the molecular events underlying sarcomatoid carcinomas (SCs) remain poorly characterised. Here, we performed targeted next-generation sequencing (NGS) on patients with surgically resected SCs comprising distinct tissues of origin. METHODS A total of 71 patients with pathological diagnosis of sarcomatoid carcinomas and underwent surgery were retrospectively enrolled in this study. Overall survival (OS) was defined as the time from surgery to death from any cause. Patients alive or lost to follow-up were censored. Genomic DNA from formalin-fixed paraffin-embedded samples was extracted for NGS and tumour mutation burden (TMB) analysis. RESULTS In general, SCs occurred more commonly in males, except those of the gallbladder. SCs of the lung and the larynx were associated with a higher proportion of smokers (p=0.0015). Alterations in TP53, RB1, TERT and KRAS were highly frequent, with KRAS mutations being a biomarker of poor prognosis (median OS=8 vs 16 months, p=0.03). Multiple alterations in potentially actionable genes, including ROS1 and NTRK1 fusions and ERBB2 amplification, were detected in the extra-pulmonary cohort. A relatively high proportion (30%) of patients with extra-pulmonary SC had high TMB, with a median of 5.39 mutations per Mb. Lastly, copy number variations were common in SCs, and were non-overlapping between the primary and metastatic tumours. CONCLUSION Taken together, our results suggest that comprehensive genetic testing may be necessary to inform treatment options and identify prognostic biomarkers.
Collapse
Affiliation(s)
- Ying Ding
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Shao
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China.,School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenglong Na
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Jiani C Yin
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Hongjin Hua
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ran Tao
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yue Jiang
- Department of Pathology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ran Hu
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao He
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Miao
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongqin Zhu
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Zhihong Zhang
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
40
|
Yao HP, Hudson R, Wang MH. Progress and challenge in development of biotherapeutics targeting MET receptor for treatment of advanced cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188425. [PMID: 32961258 DOI: 10.1016/j.bbcan.2020.188425] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022]
Abstract
Advanced epithelial cancers such as gastric, lung, and pancreatic tumors are featured by invasive proliferation, distant metastasis, acquired chemoresistance, and tumorigenic stemness. For the last decade, molecular-targeted therapies using therapeutic antibodies, small molecule kinase inhibitors and immune-checkpoint blockades have been applied for these diseases with significant clinical benefits. Nevertheless, there is still a large gap to achieve curative outcomes. MET (mesenchymal-epithelial transition protein), a receptor tyrosine kinase, is a tumorigenic determinant that regulates epithelial cancer initiation, progression, and malignancy. Increased MET expression also has prognostic value for cancer progression and patient survival. These features provide the rationale to target MET for cancer treatment. In this review, we discuss the importance of MET in epithelial tumorigenesis and the development of antibody-based biotherapeutics, including bispecific antibodies and antibody-drug conjugates, for clinical application. The findings from both preclinical and clinical studies highlight the potential of MET-targeted biotherapeutics for cancer therapy in the future.
Collapse
Affiliation(s)
- Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Rachel Hudson
- Cancer Biology Research Center, Texas Tech University Health Sciences Center, Amarillo, TX, USA; Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Biology Research Center, Texas Tech University Health Sciences Center, Amarillo, TX, USA; Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
41
|
Liu XW, Chen XR, Rong YM, Lyu N, Xu CW, Wang F, Sun WY, Fang SG, Yuan JP, Wang HJ, Wang WX, Huang WB, Xu JP, Yue ZY, Chen LK. MET exon 14 skipping mutation, amplification and overexpression in pulmonary sarcomatoid carcinoma: A multi-center study. Transl Oncol 2020; 13:100868. [PMID: 32920328 PMCID: PMC7492996 DOI: 10.1016/j.tranon.2020.100868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 01/11/2023] Open
Abstract
High frequency of MNNG HOS transforming (MET) exon 14 skipping mutation (MET exon 14Δ) has been reported in pulmonary sarcomatoid carcinomas (PSCs). However, the frequencies differ greatly. Our study aims to investigate the frequency of MET alterations and the correlations among MET exon 14Δ, amplification, and protein overexpression in a large cohort of PSCs. MET exon 14Δ, amplification, and protein overexpression were detected in 124 surgically resected PSCs by using Sanger sequencing, fluorescent in situ hybridization (FISH), and immunohistochemistry (IHC) respectively. MET exon 14Δ was identified in 9 (7.3%) of 124 cases, including 6 pleomorphic carcinomas, 2 spindle cell carcinomas and 1 carcinosarcoma. MET amplification and protein overexpression were detected in 6 PSCs (4.8%) and 25 PSCs (20.2%), respectively. MET amplification was significantly associated with overexpression (P < 0.001). However, MET exon 14Δ has no correlation with MET amplification (P = 0.370) and overexpression (P = 0.080). Multivariable analysis demonstrated that pathologic stage (hazard ratio [HR], 2.78; 95% confidence interval [CI], 1.28–6.01; P = 0.010) and MET amplification (HR, 4.71; 95% CI, 1.31–16.98; P = 0.018) were independent prognostic factors for poor median overall survival (mOS). MET alterations including MET exon 14Δ and amplification should be recommended as routine clinical testing in PSCs patients who may benefit from MET inhibitors. MET IHC appears to be an efficient screen tool for MET amplification in PSCs.
Collapse
Affiliation(s)
- Xue-Wen Liu
- Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, PR China
| | - Xin-Ru Chen
- 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, PR China
| | - Yu-Ming Rong
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Ning Lyu
- Department of Minimally Invasive Interventional Radiology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Chun-Wei Xu
- Department of Pathology, Fujian Cancer Hospital, Fujian, PR China
| | - Fang Wang
- Department of Molecular Diagnostic, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Wen-Yong Sun
- Department of Pathology, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, PR China
| | - San-Gao Fang
- Department of Pathology, Daping Hospital and Research Institute of Surgery, the Third Military Medical University, Chongqing, PR China
| | - Jing-Ping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Hui-Juan Wang
- Department of Respiratory Medicine, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Wen-Xian Wang
- Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, PR China
| | - Wen-Bin Huang
- Department of Pathology, Nanjing Hospital (Nanjing First Hospital), Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Jian-Ping Xu
- Department of Pathology, Anhui Chest Hospital, Hefei, Anhui, PR China
| | - Zhen-Ying Yue
- Dapartment of Pathology, the Central Hospital of Shengli Oilfield, Dongying, PR China
| | - Li-Kun Chen
- 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, PR China.
| |
Collapse
|
42
|
Recondo G, Che J, Jänne PA, Awad MM. Targeting MET Dysregulation in Cancer. Cancer Discov 2020; 10:922-934. [PMID: 32532746 PMCID: PMC7781009 DOI: 10.1158/2159-8290.cd-19-1446] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/31/2020] [Accepted: 04/16/2020] [Indexed: 11/16/2022]
Abstract
Aberrant MET signaling can drive tumorigenesis in several cancer types through a variety of molecular mechanisms including MET gene amplification, mutation, rearrangement, and overexpression. Improvements in biomarker discovery and testing have more recently enabled the selection of patients with MET-dependent cancers for treatment with potent, specific, and novel MET-targeting therapies. We review the known oncologic processes that activate MET, discuss therapeutic strategies for MET-dependent malignancies, and highlight emerging challenges in acquired drug resistance in these cancers. SIGNIFICANCE: Increasing evidence supports the use of MET-targeting therapies in biomarker-selected cancers that harbor molecular alterations in MET. Diverse mechanisms of resistance to MET inhibitors will require the development of novel strategies to delay and overcome drug resistance.
Collapse
Affiliation(s)
- Gonzalo Recondo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
43
|
Guo R, Luo J, Chang J, Rekhtman N, Arcila M, Drilon A. MET-dependent solid tumours - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol 2020; 17:569-587. [PMID: 32514147 DOI: 10.1038/s41571-020-0377-z] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Attempts to develop MET-targeted therapies have historically focused on MET-expressing cancers, with limited success. Thus, MET expression in the absence of a genomic marker of MET dependence is a poor predictor of benefit from MET-targeted therapy. However, owing to the development of more sensitive methods of detecting genomic alterations, high-level MET amplification and activating MET mutations or fusions are all now known to be drivers of oncogenesis. MET mutations include those affecting the kinase or extracellular domains and those that result in exon 14 skipping. The activity of MET tyrosine kinase inhibitors varies by MET alteration category. The likelihood of benefit from MET-targeted therapies increases with increasing levels of MET amplification, although no consensus exists on the optimal diagnostic cut-off point for MET copy number gains identified using fluorescence in situ hybridization and, in particular, next-generation sequencing. Several agents targeting exon 14 skipping alterations are currently in clinical development, with promising data available from early-phase trials. By contrast, the therapeutic implications of MET fusions remain underexplored. Here we summarize and evaluate the utility of various diagnostic techniques and the roles of different classes of MET-targeted therapies in cancers with MET amplification, mutation and fusion, and MET overexpression.
Collapse
Affiliation(s)
- Robin Guo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jia Luo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
44
|
Liu T, Zhao X, Zheng X, Zheng Y, Dong X, Zhao N, Liao S, Sun B. The EMT transcription factor, Twist1, as a novel therapeutic target for pulmonary sarcomatoid carcinomas. Int J Oncol 2020; 56:750-760. [PMID: 32124963 PMCID: PMC7010216 DOI: 10.3892/ijo.2020.4972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/12/2019] [Indexed: 12/25/2022] Open
Abstract
Pulmonary sarcomatoid carcinomas (PSCs) are a rare subtype of non‑small‑cell lung cancer and are typically biphasic neoplasms. No effective treatment for PSCs is currently available in clinical practice. The expression of the epithelial‑mesenchymal transition (EMT) transcription factors, Twist1, Slug and Snail, as well as the EMT phenotype and vasculogenic mimicry (VM) were analysed in 41 PSC and 79 pulmonary squamous carcinoma (PSCC) samples. Compared with the PSCCs, the PSCs exhibited an EMT phenotype and VM, and they also exhibited an increased expression of the Twist1, Slug, Snail and VM markers. Twist1 expression was associated with metastasis and TNM stage. Twist1‑positive patients exhibited a poorer prognosis for overall survival (OS) than those with Twist1‑negative PSCs. Transforming growth factor β1 (TGFβ1) was used to induce an EMT transition in a PSCC cell line. SK‑MES‑1 cells treated with TGFβ1 exhibited an increased expression of Twist1. The EMT phenotype, VM and increased migratory and invasive abilities were induced following TGFβ1 treatment. Importantly, in cells treated with TGFβ1, the EMT phenotype was reversed, VM marker expression was decreased, and the migratory and invasive ability of the PSCC cell line was decreased following Twist1 knockdown. Collectively, this study provides a new perspective of Twist1 in the aggressiveness of PSCs. The identification of Twist1 as an independent marker of poor prognoses may lead to the development of novel strategies for improving the treatment of patients with PSC.
Collapse
Affiliation(s)
- Tieju Liu
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xu Zheng
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yanjun Zheng
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Shihan Liao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
45
|
Baldacci S, Figeac M, Antoine M, Descarpentries C, Kherrouche Z, Jamme P, Copin MC, Tulasne D, Nanni I, Beau-Faller M, Melaabi S, Levallet G, Quoix E, Moro-Sibilot D, Friard S, Missy P, Barlesi F, Cadranel J, Cortot AB. High MET Overexpression Does Not Predict the presence of MET exon 14 Splice Mutations in NSCLC: Results From the IFCT PREDICT.amm study. J Thorac Oncol 2019; 15:120-124. [PMID: 31605799 DOI: 10.1016/j.jtho.2019.09.196] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 01/20/2023]
Abstract
INTRODUCTION MET proto-oncogene (MET) exon 14 splice site (METex14) mutations were recently described in NSCLC and has been reported to correlate with efficacy of MET tyrosine kinase inhibitors. High diversity of these alterations makes them hard to detect by DNA sequencing in clinical practice. Because METex14 mutations induce increased stabilization of the MET receptor, it is anticipated that these mutations are associated with MET overexpression. We aim to determine whether NSCLC with high MET overexpression could define a subset of patients with a high rate of METex14 mutations. METHODS From The French Cooperative Thoracic Intergroup PREDICT.amm cohort of 843 consecutive patients with a treatment-naive advanced NSCLC who were eligible for a first-line therapy, 108 NSCLC samples with high MET overexpression defined by an immunochemistry score 3+ were tested for METex14 mutations using fragment length analysis combined with optimized targeted next-generation sequencing. MET copy number analysis was also derived from the sequencing data. RESULTS METex14 mutations were detected in two patients (2.2%) who also displayed a TP53 mutation and a PIK3CA mutation, respectively. An MET gene copy number increase was observed in seven additional patients (7.7%). Next-generation sequencing analysis revealed inactivating mutations in TP53 (52.7%) and PTEN (1.1%), and oncogenic mutations in KRAS (28.6%), EGFR (7.7%), PIK3CA (4.4%), BRAF (4.4%), NRAS (2.2%), GNAS (1.1%), and IDH1 (1.1%). CONCLUSIONS The rate of METex14 mutations in NSCLC with high MET overexpression was similar to that found in unselected NSCLC. Moreover, we observed a high frequency of driver alterations in other oncogenes. Consequently these findings do not support the use of MET immunohistochemistry as a surrogate marker for METex14 mutations.
Collapse
Affiliation(s)
- Simon Baldacci
- Lille University Hospital, CHU Lille, Thoracic Oncology Department, Lille, France
| | - Martin Figeac
- University Lille, Functional and Structural Platform, CHU Lille, Lille, France
| | - Martine Antoine
- Department of Pathology, Hospital Tenon, AP-HP, Paris, France
| | - Clotilde Descarpentries
- Department of Biochemistry and Molecular Biology, Hormonology Metabolism Nutrition Oncology, CHU Lille, Lille, France
| | - Zoulika Kherrouche
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Philippe Jamme
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Marie-Christine Copin
- University Lille, CHU Lille, Institute of Pathology, UMR8161 CNRS, Institute of Biology of Lille, F-59000 Lille, France
| | - David Tulasne
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Isabelle Nanni
- Department of Molecular Oncology, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Michèle Beau-Faller
- Department of Molecular Biology, Strasbourg University Hospital, Strasbourg, France
| | - Samia Melaabi
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | | | - Elisabeth Quoix
- Department of Pneumology, Nouvel Hôpital Civil, University Hospital, Strasbourg, France
| | - Denis Moro-Sibilot
- Department of Pneumology, Grenoble University Hospital, Grenoble, France
| | - Sylvie Friard
- Pneumology Department, Foch Hospital, Suresnes, France
| | - Pascale Missy
- Clinical Research Unit, French Cooperative Thoracic Intergroup (IFCT), Paris, France
| | - Fabrice Barlesi
- Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Multidisciplinary Oncology & Therapeutic Innovations Department, Marseille, France
| | - Jacques Cadranel
- Chest Department-Thoracic Oncology Expert Center, AP-HP, Groupe Hospitalier HUEP, Hopital Tenon, Paris, France, and Sorbonne University, Paris, France
| | - Alexis B Cortot
- Lille University Hospital, CHU Lille, Thoracic Oncology Department, Lille, France.
| |
Collapse
|
46
|
Guo R, Berry LD, Aisner DL, Sheren J, Boyle T, Bunn PA, Johnson BE, Kwiatkowski DJ, Drilon A, Sholl LM, Kris MG. MET IHC Is a Poor Screen for MET Amplification or MET Exon 14 Mutations in Lung Adenocarcinomas: Data from a Tri-Institutional Cohort of the Lung Cancer Mutation Consortium. J Thorac Oncol 2019; 14:1666-1671. [PMID: 31228623 DOI: 10.1016/j.jtho.2019.06.009] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/04/2019] [Accepted: 06/07/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION MNNG HOS Transforming gene (MET) amplification and MET exon 14 (METex14) alterations in lung cancers affect sensitivity to MET proto-oncogene, receptor tyrosine kinase (MET [also known by the alias hepatocyte growth factor receptor]) inhibitors. Fluorescence in situ hybridization (FISH), next-generation sequencing (NGS), and immunohistochemistry (IHC) have been used to evaluate MET dependency. Here, we have determined the association of MET IHC with METex14 mutations and MET amplification. METHODS We collected data on a tri-institutional cohort from the Lung Cancer Mutation Consortium. All patients had metastatic lung adenocarcinomas and no prior targeted therapies. MET IHC positivity was defined by an H-score of 200 or higher using SP44 antibody. MET amplification was defined by copy number fold change of 1.8x or more with use of NGS or a MET-to-centromere of chromosome 7 ratio greater than 2.2 with use of FISH. RESULTS We tested tissue from 181 patients for MET IHC, MET amplification, and METex14 mutations. Overall, 71 of 181 patients (39%) were MET IHC-positive, three of 181 (2%) were MET-amplified, and two of 181 (1%) harbored METex14 mutations. Of the MET-amplified cases, two were FISH positive with MET-to-centromere of chromosome 7 ratios of 3.1 and 3.3, one case was NGS positive with a fold change of 4.4x, and one of the three cases was MET IHC-positive. Of the 71 IHC-positive cases, one (1%) was MET-amplified and two (3%) were METex14-mutated. Of the MET IHC-negative cases, two of 110 (2%) were MET-amplified. CONCLUSIONS In this study, nearly all MET IHC-positive cases were negative for MET amplification or METex14 mutations. MET IHC can also miss patients with MET amplification. The limited number of MET-amplified cases in this cohort makes it challenging to demonstrate an association between MET IHC and MET amplification. Nevertheless, IHC appears to be an inefficient screen for these genomic changes. MET amplification or METex14 mutations can best be detected by FISH and a multiplex NGS panel.
Collapse
Affiliation(s)
- Robin Guo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Lynne D Berry
- Center for Quantitative Sciences, Vanderbilt University, Nashville, Tennessee
| | - Dara L Aisner
- Department of Pathology, University of Colorado, Aurora, Colorado
| | - Jamie Sheren
- Department of Pathology, University of Colorado, Aurora, Colorado
| | - Theresa Boyle
- Department of Molecular Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Paul A Bunn
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, Colorado
| | - Bruce E Johnson
- Department of Medical Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mark G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| |
Collapse
|
47
|
O'Brien O, Wright MC, O'Brien C, Geoghegan O, Leonard N, Nicholson S, Cuffe S, Fabre A, Jochum W, Joerger M, Gray SG, Finn SP. Cost-Efficient and Easy to Perform PCR-Based Assay to Identify Met Exon 14 Skipping in Formalin-Fixed Paraffin-Embedded (FFPE) Non-Small Cell Lung Cancer (NSCLC) Samples. Diagnostics (Basel) 2019; 9:diagnostics9010013. [PMID: 30669306 PMCID: PMC6468531 DOI: 10.3390/diagnostics9010013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/10/2019] [Accepted: 01/14/2019] [Indexed: 01/03/2023] Open
Abstract
MET is a receptor tyrosine kinase (RTK) that plays important roles in carcinogenesis. Despite being frequently overexpressed in cancer, clinical responses to targeting this receptor have been limited. Recently novel splicing mutations involving the loss of exon 14 (called METex14 skipping) have emerged as potential biomarkers to predict for responsiveness to targeted therapies with Met inhibitors in non-small cell lung cancer (NSCLC). Currently, the diverse genomic alterations responsible for METex14 skipping pose a challenge for routine clinical diagnostic testing. In this report, we examine three different methodologies to detect METex14 and assess their potential utility for use as a diagnostic assay for both the identification of METex14 and intra-tumoural distribution in NSCLC.
Collapse
Affiliation(s)
- Odharnaith O'Brien
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Mark C Wright
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Cathal O'Brien
- Cancer Molecular Diagnostics, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Orla Geoghegan
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
| | - Niamh Leonard
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Siobhan Nicholson
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Sinéad Cuffe
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- HOPE Directorate, St. James's Hospital, D08 RT2X Dublin, Ireland.
| | - Aurelie Fabre
- Department of Pathology, St. Vincent's University Hospital, University College Dublin School of Medicine, D04 T6F4 Dublin, Ireland.
| | - Wolfram Jochum
- Department of Pathology, Cantonal Hospital, 9007 St. Gallen, Switzerland.
| | - Markus Joerger
- Department of Medical Oncology & Hematology, Cantonal Hospital, 9007 St. Gallen, Switzerland.
| | - Steven G Gray
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- Department of Clinical Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland.
- School of Biological Sciences, Dublin Institute of Technology, D08 NF82 Dublin, Ireland.
| | - Stephen P Finn
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
- Cancer Molecular Diagnostics, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, D08 X4RX Dublin, Ireland.
| |
Collapse
|