1
|
Zhang Q, Chen K, Yu X, Fan Y. Spotlight on the treatment of non-small cell lung cancer with rare genetic alterations and brain metastasis: Current status and future perspectives. Int J Cancer 2024; 155:2117-2128. [PMID: 38958227 DOI: 10.1002/ijc.35070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
In patients with non-small cell lung cancer (NSCLC), oncogenic variants present in <5% of cases are considered rare, the predominant of which include human epidermal growth factor receptor 2 (HER2) mutations, mesenchymal-epithelial transition (MET) alterations, c-ros oncogene 1 (ROS1) rearrangements, rearrangement during transfection (RET) fusions, v-raf mouse sarcoma virus oncogene homolog B1 (BRAF) mutations, and neurotrophic troponin receptor kinase (NTRK) fusions. Brain metastases (BMs) occur in approximately 10%-50% of patients with NSCLC harboring rare genetic variants. The recent advent of small-molecule tyrosine kinase inhibitors and macromolecular antibody-drug conjugates (ADCs) has conferred marked survival benefits to patients with NSCLC harboring rare driver alterations. Despite effective brain lesion control for most targeted agents and promising reports of intracranial remission associated with novel ADCs, BM continues to be a major therapeutic challenge. This review discusses the recent advances in the treatment of NSCLC with rare genetic variants and BM, with a particular focus on intracranial efficacy, and explores future perspectives on how best to treat these patients.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Kaiyan Chen
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaoqing Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Wang K, Leng X, Yi H, Zhang G, Hu Z, Mao Y. Lung Cancer Associated with Cystic Airspaces: Current Insights into Diagnosis, Pathophysiology, and Treatment Strategies. Cancers (Basel) 2024; 16:3930. [PMID: 39682119 DOI: 10.3390/cancers16233930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Lung cancer associated with cystic airspaces (LCCA) is a rare subtype of non-small-cell lung cancer (NSCLC), accounting for 1-4% of cases. LCCA is characterized by the presence of cystic airspaces within or at the periphery of the tumor on imaging. LCCA poses significant clinical challenges due to its high risk of misdiagnosis or missed diagnosis, often leading to a worse prognosis compared to other forms of lung cancer. While previous studies have identified correlations between the pathological features and imaging characteristics of LCCA, research on its associated driver gene mutations and responses to chemotherapy and immunotherapy remains limited. Furthermore, the development of an appropriate T-staging system is necessary to improve prognostic outcomes. This review provides an overview of the current research on the definition, imaging classification, pathological and molecular mechanisms, and prognosis of LCCA, aiming to provide a reference for clinical decision-making.
Collapse
Affiliation(s)
- Kun 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 100021, China
| | - Xuechun Leng
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital, Nanjing Medical University, Huai'an 223300, China
| | - Hang Yi
- 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 100021, China
| | - Guochao Zhang
- 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 100021, China
| | - Zhongwu Hu
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital, Nanjing Medical University, Huai'an 223300, 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 100021, China
| |
Collapse
|
3
|
Yu L, Yang R, Long Z, Tao Q, Liu B. Targeted therapy of non-small cell lung cancer: mechanisms and clinical trials. Front Oncol 2024; 14:1451230. [PMID: 39391239 PMCID: PMC11464343 DOI: 10.3389/fonc.2024.1451230] [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: 06/18/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Lung cancer is a leading cause of cancer-related deaths globally, and traditional chemotherapy has limited efficacy in treating advanced non-small cell lung cancer (NSCLC). In recent years, the prognosis for patients with NSCLC has significantly improved due to the development of new treatment modalities, including targeted therapies. Targeted therapies utilize monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), or small molecule tyrosine kinase inhibitors (TKIs) directed against specific mutated genes such as EGFR and ALK. The development of these drugs has deepened our understanding of NSCLC and improved treatment outcomes for patients. This review aims to summarize the mechanisms and current status of targeted therapy for NSCLC, discuss strategies to overcome acquired resistance, and address current challenges in the field.
Collapse
Affiliation(s)
- Le Yu
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ruoyi Yang
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zeng Long
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qingxiu Tao
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bin Liu
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Miao L, Qiu T, Li Y, Li J, Jiang X, Liu M, Zhang X, Jiang J, Zhang H, Wang Y, Li X, Ying J, Li M. Predicting MET exon 14 skipping mutation in pulmonary sarcomatoid carcinoma by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography features. Transl Lung Cancer Res 2024; 13:1232-1246. [PMID: 38973946 PMCID: PMC11225047 DOI: 10.21037/tlcr-24-56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/07/2024] [Indexed: 07/09/2024]
Abstract
Background Pulmonary sarcomatoid carcinoma (PSC) is a rare, highly malignant type of non-small cell lung cancer (NSCLC) with a poor prognosis. Targeted drugs for MET exon 14 (METex14) skipping mutation can have considerable clinical benefits. This study aimed to predict METex14 skipping mutation in PSC patients by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography (CECT) features. Methods This retrospective study included 56 patients with PSC diagnosed by pathology. All patients underwent CECT before surgery or other treatment, and both targeted DNA- and RNA-based next-generation sequencing (NGS) were used to detect METex14 skipping mutation status. The patients were divided into two groups: METex14 skipping mutation and nonmutation groups. Overall, 1,316 texture features of the whole tumour were extracted. We also collected 12 clinical and 20 conventional CECT features. After dimensionality reduction and selection, predictive models were established by multivariate logistic regression analysis. Models were evaluated using the area under the curve (AUC), and the clinical utility of the model was assessed by decision curve analysis. Results METex14 skipping mutation was detected in 17.9% of PSCs. Mutations were found more frequently in those (I) who had smaller long- or short-axis diameters (P=0.02, P=0.01); (II) who had lower T stages (I, II) (P=0.02); and (III) with pseudocapsular or annular enhancement (P=0.03). The combined model based on the conventional and texture models yielded the best performance in predicting METex14 skipping mutation with the highest AUC (0.89). The conventional and texture models also had good performance (AUC =0.83 conventional; =0.88 texture). Conclusions Whole-tumour texture analysis combined with clinical and conventional CECT features may serve as a noninvasive tool to predict the METex14 skipping mutation status in PSC.
Collapse
Affiliation(s)
- Lei Miao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Li
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Jiang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengwen Liu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiuming Jiang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huanhuan Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmei Wang
- GE Healthcare China, Pudong New Area, Shanghai, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Li
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Luo NY, Minne RL, Gallant JP, Gunaratne GS, West JL, Javeri S, Robertson AJ, Lake EW, Engle JW, Mixdorf JC, Aluicio-Sarduy E, Nickel KP, Hernandez R, Kimple RJ, Baschnagel AM, LeBeau AM. Development of an Engineered Single-Domain Antibody for Targeting MET in Non-Small Cell Lung Cancer. Bioconjug Chem 2024; 35:389-399. [PMID: 38470611 DOI: 10.1021/acs.bioconjchem.4c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
The Mesenchymal Epithelial Transition (MET) receptor tyrosine kinase is upregulated or mutated in 5% of non-small-cell lung cancer (NSCLC) patients and overexpressed in multiple other cancers. We sought to develop a novel single-domain camelid antibody with high affinity for MET that could be used to deliver conjugated payloads to MET expressing cancers. From a naïve camelid variable-heavy-heavy (VHH) domain phage display library, we identified a VHH clone termed 1E7 that displayed high affinity for human MET and was cross-reactive with MET across multiple species. When expressed as a bivalent human Fc fusion protein, 1E7-Fc was found to selectively bind to EBC-1 (MET amplified) and UW-Lung 21 (MET exon 14 mutated) cell lines by flow cytometry and immunofluorescence imaging. Next, we investigated the ability of [89Zr]Zr-1E7-Fc to detect MET expression in vivo by PET/CT imaging. [89Zr]Zr-1E7-Fc demonstrated rapid localization and high tumor uptake in both xenografts with a %ID/g of 6.4 and 5.8 for EBC-1 and UW-Lung 21 at 24 h, respectively. At the 24 h time point, clearance from secondary and nontarget tissues was also observed. Altogether, our data suggest that 1E7-Fc represents a platform technology that can be employed to potentially both image and treat MET-altered NSCLC.
Collapse
Affiliation(s)
- Natalie Y Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Rachel L Minne
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Joseph P Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Gihan S Gunaratne
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Jayden L West
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Saahil Javeri
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Austin J Robertson
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Eric W Lake
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Jonathan W Engle
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Jason C Mixdorf
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Eduardo Aluicio-Sarduy
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Kwang P Nickel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Reinier Hernandez
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Randall J Kimple
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Andrew M Baschnagel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| |
Collapse
|
6
|
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: 9] [Impact Index Per Article: 9.0] [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
|
7
|
Mendes Serrão E, Klug M, Moloney BM, Jhaveri A, Lo Gullo R, Pinker K, Luker G, Haider MA, Shinagare AB, Liu X. Current Status of Cancer Genomics and Imaging Phenotypes: What Radiologists Need to Know. Radiol Imaging Cancer 2023; 5:e220153. [PMID: 37921555 DOI: 10.1148/rycan.220153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Ongoing discoveries in cancer genomics and epigenomics have revolutionized clinical oncology and precision health care. This knowledge provides unprecedented insights into tumor biology and heterogeneity within a single tumor, among primary and metastatic lesions, and among patients with the same histologic type of cancer. Large-scale genomic sequencing studies also sparked the development of new tumor classifications, biomarkers, and targeted therapies. Because of the central role of imaging in cancer diagnosis and therapy, radiologists need to be familiar with the basic concepts of genomics, which are now becoming the new norm in oncologic clinical practice. By incorporating these concepts into clinical practice, radiologists can make their imaging interpretations more meaningful and specific, facilitate multidisciplinary clinical dialogue and interventions, and provide better patient-centric care. This review article highlights basic concepts of genomics and epigenomics, reviews the most common genetic alterations in cancer, and discusses the implications of these concepts on imaging by organ system in a case-based manner. This information will help stimulate new innovations in imaging research, accelerate the development and validation of new imaging biomarkers, and motivate efforts to bring new molecular and functional imaging methods to clinical radiology. Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
- Eva Mendes Serrão
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Maximiliano Klug
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Brian M Moloney
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Aaditeya Jhaveri
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Roberto Lo Gullo
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Katja Pinker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Gary Luker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Masoom A Haider
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Atul B Shinagare
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Xiaoyang Liu
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| |
Collapse
|
8
|
Gow CH, Hsieh MS, Chen YL, Liu YN, Wu SG, Shih JY. Survival outcomes and prognostic factors of lung cancer patients with the MET exon 14 skipping mutation: A single-center real-world study. Front Oncol 2023; 13:1113696. [PMID: 36969059 PMCID: PMC10034335 DOI: 10.3389/fonc.2023.1113696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionThe MET exon 14 skipping (METex14) mutation is an important oncogenic driver in lung cancer. We performed a retrospective analysis of clinical data from lung cancer patients with the METex14 mutation to analyze their survival outcomes and associated prognostic factors.MethodsA one-step reverse transcription-polymerase chain reaction to examine the presence of the METex14 mutation was performed using RNA samples from 1374 lung cancer patients with no detected EGFR and ALK mutations. Pathological features and immunohistochemistry (IHC) results for c-MET were analyzed in patients with METex14-positive tumors.ResultsMETex14 was identified in 69 patients with lung cancer, including 53 adenocarcinoma (ADC) and 16 non-ADC patients. In comparison with patients without the METex14 mutation, lung cancer patients harboring the METex14 mutation were generally elderly individuals, never-smokers, and had poor performance scores. A higher frequency of METex14 mutations was detected in pulmonary sarcomatoid carcinoma (PSC) patients (24.3%, n = 9/37). However, stage IV PSC patients with or without the METex14 mutations showed similarly poor overall survival (OS) (p = 0.429). For all 36 METex14-positive lung ADCs, multivariate analysis showed several poor prognostic factors, including strong c-MET IHC staining (p = 0.006), initial brain metastasis (p = 0.005), and administration of only supportive care (p < 0.001). After excluding seven patients who received only supportive care, we further analyzed 29 stage IV lung ADC patients with METex14 mutations who received anti-cancer treatment. Multivariate analysis showed that pemetrexed treatment (p = 0.003), lung radiotherapy (p = 0.020), initial brain metastasis (p = 0.005), and strong c-MET IHC staining (p = 0.012) were independent prognostic factors for OS in these patients.ConclusionsA higher frequency of METex14 mutations was detected in PSC patients. Stage IV PSC patients with or without the METex14 mutations had similarly poor overall survival. Pemetrexed-based chemotherapy, strong c-MET ICH staining, initial brain metastasis, and lung radiotherapy, may help predict survival outcomes in patients with advanced lung ADCs harboring the METex14 mutation.
Collapse
Affiliation(s)
- Chien-Hung Gow
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Healthcare Information and Management, Ming-Chuan University, Taoyuan, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Lin Chen
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Nan Liu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shang-Gin Wu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Cancer Center, National Taiwan University, Taipei, Taiwan
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
- *Correspondence: Jin-Yuan Shih,
| |
Collapse
|
9
|
[Research Progresses in the Treatment of NSCLC with MET Gene Variants: A Riview]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:877-887. [PMID: 36617474 PMCID: PMC9845091 DOI: 10.3779/j.issn.1009-3419.2022.101.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal-epithelial transition factor (MET) has long been considered as the most crucial and promising driver gene in the occurrence and development of non-small cell lung cancer (NSCLC), except for epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), and c-ROS oncogene 1 receptor tyrosine kinase (ROS1). In recent years, therapeutic drugs targeting MET have been continuously developed and applied in clinical practice. First, the curative effect of NSCLC patients with MET exon 14 skipping mutations has been further improved. In addition, when MET amplification occurs after resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) in patients with advanced EGFR-mutant NSCLC, the combination of MET-TKIs and EGFR-TKIs has brought significant survival benefits and many other advances. This article reviews the treatment progress of NSCLC patients with different types of MET variants under different circumstances, which provides reference for the selection of clinical treatment strategies.
.
Collapse
|
10
|
Lai GGY, Guo R, Drilon A, Shao Weng Tan D. Refining patient selection of MET-activated non-small cell lung cancer through biomarker precision. Cancer Treat Rev 2022; 110:102444. [PMID: 36108503 PMCID: PMC10961969 DOI: 10.1016/j.ctrv.2022.102444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 12/12/2022]
Abstract
Dysregulated MET signaling plays an important role in lung oncogenesis, tumor growth and invasiveness. It may occur through various mechanisms, such as MET overexpression or gene amplification or mutation, all of which can be detected by specific methods. The utility of MET overexpression as a biomarker remains unclear due to discrepancies in its occurrence and non-standardized cut-off thresholds. MET exon 14 skipping mutation (METex14) was established as a strong predictor of response to selective MET tyrosine kinase inhibitors (TKIs), and clinical trial results in patients with non-small cell lung cancer (NSCLC) harboring METex14 led to the approval of capmatinib and tepotinib by regulatory agencies worldwide. MET amplification is an emerging biomarker, with clinical data indicating an association between MET gene copy number and response to MET-TKIs. Additionally, MET amplification represents an important mechanism of resistance to TKIs in oncogene-driven NSCLC. The identification of molecular alterations for which targeted therapies are available is important, and high-throughput next-generation sequencing techniques can provide information on multiple genes at the same time, helping to provide valuable predictive information for oncogene-driven cancers. This review summarizes the current methods used for the detection of METex14, MET amplification and MET overexpression, and discusses the evidence for the use of MET-TKIs in patients with NSCLC with MET dysregulation. We discuss the practical challenges that impact the use of METex14 in the clinic and the evidence gaps that need to be addressed to validate additional genomic markers for MET-dependent cancers.
Collapse
Affiliation(s)
- Gillianne G Y Lai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Robin Guo
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
11
|
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: 56] [Impact Index Per Article: 18.7] [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
|
12
|
Characteristic computed tomography features in mesenchymal-epithelial transition exon14 skipping-positive non-small cell lung cancer. BMC Pulm Med 2022; 22:260. [PMID: 35773658 PMCID: PMC9245203 DOI: 10.1186/s12890-022-02037-4] [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: 03/31/2022] [Accepted: 06/13/2022] [Indexed: 11/18/2022] Open
Abstract
Background Mesenchymal-epithelial transition exon14 (METex14) skipping is one of the therapeutic driver oncogene mutations in non-small cell lung cancer (NSCLC), and can be treated with tepotinib and capmatinib. There is only one report on computed tomography (CT) findings of METex14 skipping-positive NSCLC, which shows that the primary tumor tends to have a large mass in the upper lobe, and extrathoracic metastases are common. This study examined the CT findings of METex14 skipping-positive NSCLC, focusing on the features of the margins and internal structures. Methods We consecutively included patients with METex14 skipping-positive NSCLC who were diagnosed between January 2018 and December 2020 at four independent institutions. We retrospectively reviewed the patient demographics and CT findings for tumor margins (invasion into surrounding tissue, lobulation, pleural indentation, spicula, and ground-glass opacity) and internal structures (air bronchograms, cavitation and internal low-density area). Results Fifteen patients with METex14 skipping-positive NSCLC were identified. Almost half of the patients were men (7/15; 46.7%), and their median age was 75.0 years. More than half were either current or former smokers (9/15; 60.0%). A vast majority of histological subtypes were adenocarcinoma (10/15; 66.7%), followed by pleomorphic carcinoma (3/15; 20.0%) and squamous cell carcinoma (2/15; 13.3%). With regard to CT findings, most primary tumors presented as masses larger than 30 mm (12/15; 80.0%) and were located in the upper lobes (12/15; 80.0%). Invasion into surrounding tissue and presence of internal low-density areas were observed in 60.0% (9/15) and 66.7% (10/15) of the primary tumors, respectively. Additionally, their frequencies increased to 72.7% (8/11) and 90.9% (10/11) in stage III/IV cases, respectively. In lymph node metastasis, internal low-density areas were observed in 8/10 cases (80.0%). Although these two CT features were rarely observed in distant metastases at diagnosis, they became apparent with progression of the metastatic tumor size. Conclusions METex14 skipping-positive NSCLC tumors tend to invade surrounding tissue and possess internal low-density areas. These CT findings might be characteristic of METex14 skipping-positive NSCLC. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-02037-4.
Collapse
|
13
|
Hamilton G, Rath B. Met inhibitors in the treatment of lung cancer: the evidence to date. Expert Opin Pharmacother 2022; 23:815-825. [PMID: 35377279 DOI: 10.1080/14656566.2022.2062227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : The hepatocyte growth factor (HGF) receptor MET is an oncogenic driver in a subpopulation of Non-small Lung Cancer Cells (NSCLC) at the primary tumor stage or in acquired resistance to treatment with tumor-targeting tyrosine kinase inhibitors (TKIs). AREAS COVERED This article summarizes the mechanisms leading to overexpression and activation of MET by amplification and mutations including exon 14 aberrations. Furthermore, the methods to detect and categorize MET as a tumor driver and the selective TKIs for patient treatment are discussed. EXPERT OPINION : Activating mutations and rearrangements of kinases in NSCLC are the target of successful therapeutic intervention. However, MET activation involves a number of complex alterations including gene amplification, prevention of degradation by METex14 exon skipping and a host of gene mutations. A high-level of MET expression is the precondition for tumor responses to TKIs and the confirmation of MET-dependent tumor progression is difficult in primary lesions and in tumors exhibiting resistance to mutated EGFR-directed therapy in absence of standardized and concordant assays of MET amplification.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Le X, Sakai H, Felip E, Veillon R, Garassino MC, Raskin J, Cortot AB, Viteri S, Mazieres J, Smit EF, Thomas M, Iams WT, Cho BC, Kim HR, Yang JCH, Chen YM, Patel JD, Bestvina CM, Park K, Griesinger F, Johnson M, Gottfried M, Britschgi C, Heymach J, Sikoglu E, Berghoff K, Schumacher KM, Bruns R, Otto G, Paik PK. Tepotinib Efficacy and Safety in Patients with MET Exon 14 Skipping NSCLC: Outcomes in Patient Subgroups from the VISION Study with Relevance for Clinical Practice. Clin Cancer Res 2022; 28:1117-1126. [PMID: 34789481 PMCID: PMC9365370 DOI: 10.1158/1078-0432.ccr-21-2733] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/29/2021] [Accepted: 11/11/2021] [Indexed: 01/26/2023]
Abstract
PURPOSE Primary analysis of VISION showed tepotinib had durable clinical activity in patients with MET exon 14 (METex14) skipping non-small cell lung cancer (NSCLC). We present updated outcomes for clinically relevant subgroups. PATIENTS AND METHODS This phase II, open-label, multi-cohort study of 500 mg (450 mg active moiety) tepotinib in patients with METex14 skipping NSCLC assessed efficacy and safety in predefined subgroups according to age, prior therapies (chemotherapy and immune checkpoint inhibitors), and brain metastases. An ad hoc retrospective analysis using Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) criteria assessed intracranial activity. RESULTS 152 patients were evaluable for efficacy (median age: 73.1). Overall, objective response rate (ORR) was 44.7% [95% confidence interval (CI): 36.7-53.0]. Patients aged <75 (n = 84) and ≥75 (n = 68) had ORRs of 48.8% (95% CI: 37.7-60.0) and 39.7% (95% CI: 28.0-52.3), respectively. Treatment-naïve (n = 69) versus previously treated (n = 83) patients showed consistent efficacy [ORR (95% CI): 44.9% (32.9-57.4) vs. 44.6% (33.7-55.9); median duration of response (95% CI): 10.8 (6.9-not estimable) vs. 11.1 (9.5-18.5) months]. Of 15 patients analyzed by RANO-BM (12 received prior radiotherapy), 13 achieved intracranial disease control; 5 of 7 patients with measurable brain metastases had partial intracranial responses. Of 255 patients evaluable for safety, 64 (25.1%) experienced grade ≥3 treatment-related adverse events (TRAE), leading to discontinuation in 27 patients (10.6%). Rates of adverse events (AE) were broadly consistent irrespective of prior therapies. CONCLUSIONS Tepotinib showed meaningful activity across subgroups by age, prior therapies, and brain metastases, with a manageable safety profile and few treatment discontinuations. See related commentary by Rosner and Spira, p. 1055.
Collapse
Affiliation(s)
- Xiuning Le
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Corresponding Author: Xiuning Le, Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: 713-792-6363; E-mail:
| | - Hiroshi Sakai
- Department of Thoracic Oncology, Saitama Cancer Center, Kitaadachi-gun, Japan
| | - Enriqueta Felip
- Department of Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Remi Veillon
- CHU Bordeaux, Service des Maladies Respiratoires, Bordeaux, France
| | - Marina Chiara Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Medicine, Section of Hematology/Oncology, Knapp Center for Biomedical Discovery, The University of Chicago, Chicago, Illinois
| | - Jo Raskin
- Department of Pulmonology and Thoracic Oncology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Alexis B. Cortot
- Univ. Lille, CHU Lille, CNRS, Inserm, Institut Pasteur de Lille, UMR9020 – UMR-S 1277 - Canther, Lille, France
| | - Santiago Viteri
- Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Grupo Quiron Salud, Barcelona, Spain
| | - Julien Mazieres
- CHU de Toulouse, Institut Universitaire du Cancer, Toulouse, France
| | - Egbert F. Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michael Thomas
- Thoraxklinik, University Heidelberg and Translational Lung Research Center Heidelberg (TLRC-H), The German Center for Lung Research (DZL), Heidelberg, Germany
| | - Wade T. Iams
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Ryun Kim
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - James Chih-Hsin Yang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, and School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jyoti D. Patel
- Lurie Cancer Center, Northwestern University-Feinberg School of Medicine, Chicago, Illinois
| | | | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Frank Griesinger
- Pius-Hospital, University Medicine Oldenburg, Department of Hematology and Oncology, University Department Internal Medicine-Oncology, Oldenburg, Germany
| | - Melissa Johnson
- Department of Medicine, Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Maya Gottfried
- Department of Oncology, Meir Medical Center, Tchernichovsky St 59, Kefar Sava, Israel
| | - Christian Britschgi
- Department of Medical Oncology and Hematology, Comprehensive Cancer Center Zurich, University Hospital Zurich, Zurich, Switzerland
| | - John Heymach
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elif Sikoglu
- Calyx, Patient Technology Solutions, Medical Imaging, Billerica, Massachusetts
| | - Karin Berghoff
- Global Patient Safety, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Karl-Maria Schumacher
- Global Clinical Development, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Rolf Bruns
- Department of Biostatistics, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Gordon Otto
- Global Clinical Development, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Paul K. Paik
- Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
15
|
Terlecka P, Krawczyk P, Grenda A, Milanowski J. MET Gene Dysregulation as a Promising Therapeutic Target in Lung Cancer-A Review. J Pers Med 2021; 11:1370. [PMID: 34945842 PMCID: PMC8705301 DOI: 10.3390/jpm11121370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/20/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Several molecular abnormalities in the MET gene have been identified, including overexpression, amplification, point mutations, and "skipping mutation" in exon 14. Even though deregulated MET signaling occurs rarely in non-small cell lung cancer (NSCLC), it possesses tumorigenic activity. Since the discovery of the significant role played by MET dysregulations in resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKI), many clinical trials have been focused on mechanisms underlying this acquired resistance. Therefore, new therapeutic strategies are being considered in the personalized therapy of NSCLC patients carrying MET abnormalities. First, MET kinase inhibitors (tepotinib and capmatinib) have been shown to be effective in the first and subsequent lines of treatment in NSCLC patients with "skipping mutations" in exon 14 of MET gene. In this article, the authors show the role of MET signaling pathway alterations and describe the results of clinical trials with MET inhibitors in NSCLC patients.
Collapse
Affiliation(s)
- Paulina Terlecka
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (P.K.); (A.G.); (J.M.)
| | | | | | | |
Collapse
|
16
|
Brain penetration and efficacy of tepotinib in orthotopic patient-derived xenograft models of MET-driven non-small cell lung cancer brain metastases. Lung Cancer 2021; 163:77-86. [PMID: 34942492 DOI: 10.1016/j.lungcan.2021.11.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 11/20/2022]
Abstract
Central nervous system-penetrant therapies with intracranial efficacy against non-small cell lung cancer (NSCLC) brain metastases are urgently needed. We report preclinical studies investigating brain penetration and intracranial activity of the MET inhibitor tepotinib. After intravenous infusion of tepotinib in Wistar rats (n = 3), mean (±standard deviation) total tepotinib concentration was 2.87-fold higher in brain (505 ± 22 ng/g) than plasma (177 ± 20 ng/mL). In equilibrium dialysis experiments performed in triplicate, mean tepotinib unbound fraction was 0.35% at 0.3 and 3.0 µM tepotinib in rat brain tissue, and 4.0% at 0.3 and 1.0 µM tepotinib in rat plasma. The calculated unbound brain-to-plasma ratio was 0.25, indicating brain penetration sufficient for intracranial target inhibition. Of 20 screened subcutaneous patient-derived xenograft (PDX) models from lung cancer brain metastases (n = 1), two NSCLC brain metastases models (LU5349 and LU5406) were sensitive to the suboptimal dose of tepotinib of 30 mg/kg/qd (tumor volume change [%TV]: -12% and -88%, respectively). Molecular profiling (nCounter®; NanoString) revealed high-level MET amplification in both tumors (mean MET gene copy number: 11.2 and 24.2, respectively). Tepotinib sensitivity was confirmed for both subcutaneous models at a clinically relevant dose (125 mg/kg/qd; n = 5). LU5349 and LU5406 were orthotopically implanted into brains of mice and monitored by magnetic resonance imaging (MRI). Tepotinib 125 mg/kg/qd induced pronounced tumor regression, including complete or near-complete regressions, compared with vehicle in both orthotopic models (n = 10; median %TV: LU5349, -84%; LU5406, -63%). Intracranial antitumor activity of tepotinib did not appear to correlate with blood-brain barrier leakiness assessed in T1-weighted gadolinium contrast-enhanced MRI.
Collapse
|
17
|
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
|
18
|
Wu MY, Zhang EW, Strickland MR, Mendoza DP, Lipkin L, Lennerz JK, Gainor JF, Heist RS, Digumarthy SR. Clinical and Imaging Features of Non-Small Cell Lung Cancer with G12C KRAS Mutation. Cancers (Basel) 2021; 13:cancers13143572. [PMID: 34298783 PMCID: PMC8304953 DOI: 10.3390/cancers13143572] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/14/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary KRAS G12C mutations are important oncogenic mutations in lung cancer that can now be targeted by allosteric small molecule inhibitors. We assessed the imaging features and patterns of metastases in these lung cancers compared to other mutated lung cancers. We found that KRAS G12C NSCLC has distinct primary tumor imaging features and patterns of metastasis when compared to those of NSCLC driven by other genetic alterations. These distinct imaging features may offer clues to its presence and potentially guide management in the future. Abstract KRAS G12C mutations are important oncogenic mutations that confer sensitivity to direct G12C inhibitors. We retrospectively identified patients with KRAS+ NSCLC from 2015 to 2019 and assessed the imaging features of the primary tumor and the distribution of metastases of G12C NSCLC compared to those of non-G12C KRAS NSCLC and NSCLC driven by oncogenic fusion events (RET, ALK, ROS1) and EGFR mutations at the time of initial diagnosis. Two hundred fifteen patients with KRAS+ NSCLC (G12C: 83; non-G12C: 132) were included. On single variate analysis, the G12C group was more likely than the non-G12C KRAS group to have cavitation (13% vs. 5%, p = 0.04) and lung metastasis (38% vs. 21%; p = 0.043). Compared to the fusion rearrangement group, the G12C group had a lower frequency of pleural metastasis (21% vs. 41%, p = 0.01) and lymphangitic carcinomatosis (4% vs. 39%, p = 0.0001) and a higher frequency of brain metastasis (42% vs. 22%, p = 0.005). Compared to the EGFR+ group, the G12C group had a lower frequency of lung metastasis (38% vs. 67%, p = 0.0008) and a higher frequency of distant nodal metastasis (10% vs. 2%, p = 0.02). KRAS G12C NSCLC may have distinct primary tumor imaging features and patterns of metastasis when compared to those of NSCLC driven by other genetic alterations.
Collapse
Affiliation(s)
- Markus Y. Wu
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA 02114, USA; (M.Y.W.); (E.W.Z.); (D.P.M.)
| | - Eric W. Zhang
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA 02114, USA; (M.Y.W.); (E.W.Z.); (D.P.M.)
| | - Matthew R. Strickland
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (M.R.S.); (J.F.G.); (R.S.H.)
| | - Dexter P. Mendoza
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA 02114, USA; (M.Y.W.); (E.W.Z.); (D.P.M.)
| | - Lev Lipkin
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; (L.L.); (J.K.L.)
| | - Jochen K. Lennerz
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; (L.L.); (J.K.L.)
| | - Justin F. Gainor
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (M.R.S.); (J.F.G.); (R.S.H.)
| | - Rebecca S. Heist
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (M.R.S.); (J.F.G.); (R.S.H.)
| | - Subba R. Digumarthy
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA 02114, USA; (M.Y.W.); (E.W.Z.); (D.P.M.)
- Correspondence: ; Tel.: +1-617-724-4254; Fax: +1-617-724-0046
| |
Collapse
|
19
|
Bittoni M, Yang JCH, Shih JY, Peled N, Smit EF, Camidge DR, Arasada RR, Oksen D, Boutmy E, Stroh C, Johne A, Carbone DP, Paik PK. Real-world insights into patients with advanced NSCLC and MET alterations. Lung Cancer 2021; 159:96-106. [PMID: 34320421 PMCID: PMC9345068 DOI: 10.1016/j.lungcan.2021.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/16/2022]
Abstract
Objectives: To describe characteristics, treatment and outcomes of non-small cell lung cancer (NSCLC) patients with MET alterations (MET exon 14 [METex14] skipping or MET amplification [METamp]) in real-world clinical care. Methods: This non-interventional cohort study used real-world data extracted from electronic medical records from academic oncology sites in Israel, The Netherlands, Taiwan, and the USA. Patients had confirmed diagnosis of advanced (Stage IIIB–IV) NSCLC harboring MET alterations (date of diagnosis = index date) between 1 Jan 2010 and 30 Sept 2018. Medical history was assessed prior to and at the index date (baseline period), and outcomes from first date of treatment to death, loss to follow-up, or end of study period. Results: A total of 117 patients were included (METex14 n = 70; METamp n = 47); testing methods were heterogeneous. Concomitant oncogenic mutations were more common in the METamp cohort than METex14. Patients in the METex14 cohort were older than those in METamp, and a larger proportion were never smokers. Anticancer first-line therapies received by patients (METex14; METamp) included chemotherapy only (44%; 41%), MET inhibitors (33%; 29%), immune checkpoint inhibitor (ICI) mono-(12%; 15%) and combination-therapy (8%; 3%). Second-line therapies included chemotherapy (35%; 30%) and MET inhibitors (30%; 39%). In the METex14 cohort, objective response rate (ORR) was generally low (first-line 28%; second-line 30%); no patients who received ICIs had a response. In the METamp cohort, ORR was 36% in first-line and 22% in second-line. Median (95% confidence interval) overall survival from start of first-line therapy was 12.0 months (6.8, 19.2) in the METex14 cohort and 22.0 months (9.8, 31.2) in METamp. Conclusions: Heterogeneous treatments reflect the changing landscape and availability of new treatments, as well as the high unmet medical need in older, METex14 patients who had more advanced disease at diagnosis. MET-targeted therapies could be beneficial in patients with these rare MET alterations.
Collapse
Affiliation(s)
- Marisa Bittoni
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - James Chih-Hsin Yang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taiwan.
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Nir Peled
- Oncology Department, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Egbert F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - D Ross Camidge
- Medical Oncology Department, University of Colorado, Aurora, CO, USA.
| | | | - Dina Oksen
- Research and Development, Merck Healthcare KGaA, Darmstadt, Germany.
| | - Emmanuelle Boutmy
- Research and Development, Merck Healthcare KGaA, Darmstadt, Germany.
| | - Christopher Stroh
- Research and Development, Merck Healthcare KGaA, Darmstadt, Germany.
| | - Andreas Johne
- Research and Development, Merck Healthcare KGaA, Darmstadt, Germany.
| | - David P Carbone
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Paul K Paik
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
20
|
Selecting the optimal immunotherapy regimen in driver-negative metastatic NSCLC. Nat Rev Clin Oncol 2021; 18:625-644. [PMID: 34168333 DOI: 10.1038/s41571-021-00520-1] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
The treatment landscape of driver-negative non-small-cell lung cancer (NSCLC) is rapidly evolving. Immune-checkpoint inhibitors, specifically those targeting PD-1 or PD-L1, have demonstrated durable efficacy in a subset of patients with NSCLC, and these agents have become the cornerstone of first-line therapy. Approved immunotherapeutic strategies for treatment-naive patients now include monotherapy, immunotherapy-exclusive regimens or chemotherapy-immunotherapy combinations. Decision making in this space is complex given the absence of head-to-head prospective comparisons, although a thorough analysis of long-term efficacy and safety data from pivotal clinical trials can provide insight into the optimal management of each subset of patients. Indeed, histological subtype and the extent of tumour cell PD-L1 expression are paramount to regimen selection, although other clinicopathological factors and patient preferences might also be relevant in certain scenarios. Finally, several emerging biomarkers and novel therapeutic strategies are currently under investigation, and these might further refine the current treatment paradigm. In this Review, we discuss the current treatment landscape and detail our approach to first-line immunotherapy regimen selection for patients with advanced-stage, driver-negative NSCLC.
Collapse
|
21
|
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: 0.5] [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
|
22
|
Rebuzzi SE, Zullo L, Rossi G, Grassi M, Murianni V, Tagliamento M, Prelaj A, Coco S, Longo L, Dal Bello MG, Alama A, Dellepiane C, Bennicelli E, Malapelle U, Genova C. Novel Emerging Molecular Targets in Non-Small Cell Lung Cancer. Int J Mol Sci 2021; 22:ijms22052625. [PMID: 33807876 PMCID: PMC7961376 DOI: 10.3390/ijms22052625] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
In the scenario of systemic treatment for advanced non-small cell lung cancer (NSCLC) patients, one of the most relevant breakthroughs is represented by targeted therapies. Throughout the last years, inhibitors of the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-Ros oncogene 1 (ROS1), and V-raf murine sarcoma viral oncogene homolog B (BRAF) have been approved and are currently used in clinical practice. However, other promising molecular drivers are rapidly emerging as therapeutic targets. This review aims to cover the molecular alterations with a potential clinical impact in NSCLC, including amplifications or mutations of the mesenchymal–epithelial transition factor (MET), fusions of rearranged during transfection (RET), rearrangements of the neurotrophic tyrosine kinase (NTRK) genes, mutations of the Kirsten rat sarcoma viral oncogene (KRAS) and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA), as well as amplifications or mutations of human epidermal growth factor receptor 2 (HER2). Additionally, we summarized the current status of targeted agents under investigation for such alterations. This revision of the current literature on emerging molecular targets is needed as the evolving knowledge on novel actionable oncogenic drivers and targeted agents is expected to increase the proportion of patients who will benefit from tailored therapeutic approaches.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.G.); (V.M.)
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy; (M.T.); (C.G.)
- Correspondence:
| | - Lodovica Zullo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Giovanni Rossi
- Medical Oncology Department, Ospedale Padre Antero Micone, 16153 Genoa, Italy;
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via Roma 151, 07100 Sassari, Italy
| | - Massimiliano Grassi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.G.); (V.M.)
| | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.G.); (V.M.)
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy; (M.T.); (C.G.)
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Arsela Prelaj
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
- Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Luca Longo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Angela Alama
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Chiara Dellepiane
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Elisa Bennicelli
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80138 Naples, Italy;
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy; (M.T.); (C.G.)
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
23
|
Hong L, Zhang J, Heymach JV, Le X. Current and future treatment options for MET exon 14 skipping alterations in non-small cell lung cancer. Ther Adv Med Oncol 2021; 13:1758835921992976. [PMID: 33643443 PMCID: PMC7890719 DOI: 10.1177/1758835921992976] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
It has been over three decades since the hepatocyte growth factor (HGF) ligand and its receptor MET proto-oncogene (MET) pathway was established as promoting cancer growth and metastasis. MET exon 14 skipping (METex14) alterations occur in 3-4% of all non-small cell lung cancer (NSCLC) patients, typically in elderly patients (older than 70 years), and result in constitutive activation of the MET receptor by altering a region required for receptor degradation. Multi-kinase inhibitor of MET, such as crizotinib, and more recently selective MET inhibitors, such as capmatinib and tepotinib, have demonstrated clinical efficacy and safety in METex14 NSCLC patients in clinical trials. These results have led to the approval of MET inhibitors by regulatory agencies across the globe. The success also fueled the excitement of further development of therapeutic strategies to target METex14 in lung cancers. This article provides an overview of the clinical development program targeting METex14 in NSCLC, including small molecular tyrosine kinase inhibitors and anti-MET antibodies. Furthermore, combination therapy immune checkpoint inhibitors or other targeted therapies are also under development in various patient populations, with acquired resistance immune or targeted therapy. Clinical trials in different development stages are ongoing and more drugs targeted to c-MET will be available for NSCLC patients with METex14 skipping mutations in the future.
Collapse
Affiliation(s)
- Lingzhi Hong
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
24
|
Mendoza DP, Heeger A, Mino-Kenudson M, Lanuti M, Shepard JAO, Sequist LV, Digumarthy SR. Clinicopathologic and Longitudinal Imaging Features of Lung Cancer Associated With Cystic Airspaces: A Systematic Review and Meta-Analysis. AJR Am J Roentgenol 2021; 216:318-329. [PMID: 32755209 DOI: 10.2214/ajr.20.23835] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND. Lung cancer (LC) associated with cystic airspaces is an uncommon presentation that is underrecognized on imaging. Additionally, understanding of its underlying pathology and risk factors is limited, which can contribute to delays in diagnosis. OBJECTIVE. The purpose of this analysis was to systematically review, analyze, and synthesize the medical literature to determine the imaging features of LC associated with cystic airspaces. EVIDENCE ACQUISITION. In accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, we included published research reporting the clinical, pathologic, and imaging features of LC associated with cystic airspaces. We then performed a pooled analysis of continuous and categoric data with respect to patient clinical characteristics, tumor pathologic features, underlying driver mutation, CT features, and evolution of these features over time. EVIDENCE SYNTHESIS. The analysis included eight original observational studies with a combined total of 341 patients with LC associated with cystic airspaces (weighted mean age, 61.8 years; range, 30-87 years; 135 women and 206 men). Most patients were current or previous smokers (127/192 [66.1%]). The most common histologic finding was adenocarcinoma (289/328 [88.1%]) followed by squamous cell carcinoma (30/328 [9.1%]). The most common driver mutations were EGFR (46/122 [37.7%]) and KRAS (21/122 [17.2%]). The cysts in LC associated with cystic airspaces commonly had nonuniform (104/114 [91.2%]) and thick (83/222 [37.4%]) walls, irregular margins (53/142 [37.3%]), and were unilocular (173/272 [63.6%]). Most cysts had a nodular component (210/328 [64.0%]). Over time, most cysts showed development or enlargement of the nodular component (61/89 [68.5%]), approximately half showed wall thickening (43/89 [48.3%]), and a minority evolved into completely solid lesions (11/89 [12.4%]). The size of the cystic component increased in 36 of 89 patients (40.4%), decreased in 28 (31.5%), and remained stable in 24 (27.0%). CONCLUSION. LC associated with cystic airspaces occurs most commonly as adeno-carcinoma and is seen in both smokers and nonsmokers. The cysts associated with LC show wall thickening and mural nodularity, which may evolve over time. LC associated with cystic airspaces can be indolent, and long-term surveillance with imaging should be considered if cysts are not resected. CLINICAL IMPACT. Familiarity with the imaging features and temporal evolution of LC associated with cystic airspaces can minimize delays in LC diagnosis. Future management guidelines should include protocols for follow-up and management of cystic lung lesions identified during diagnostic and LC screening CT.
Collapse
Affiliation(s)
- Dexter P Mendoza
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Founders 202, Boston, MA 02114
| | - Allen Heeger
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Founders 202, Boston, MA 02114
| | | | - Michael Lanuti
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Jo-Anne O Shepard
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Founders 202, Boston, MA 02114
| | | | - Subba R Digumarthy
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Founders 202, Boston, MA 02114
| |
Collapse
|
25
|
Management and Outcomes of Suspected Infectious and Inflammatory Lung Abnormalities Identified on Lung Cancer Screening CT. AJR Am J Roentgenol 2020; 217:1083-1092. [PMID: 33377416 DOI: 10.2214/ajr.20.25124] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background: Incidental findings are frequently encountered during lung cancer screening (LCS). Limited data describe the prevalence of suspected acute infectious and inflammatory lung processes on LCS and how they should be managed. Objective: To determine the prevalence, radiologic reporting and management, and outcome of suspected infectious and inflammatory lung processes identified incidentally during LCS, and to propose a management algorithm. Methods: This retrospective study included 6314 low dose CT (LDCT) examinations performed between June 2014 and April 2019 in 3800 patients as part of an established LCS program. Radiology reports were reviewed, and patients with potentially infectious or inflammatory lung abnormalities were identified and analyzed for descriptors of imaging findings, Lung-RADS designation, recommendations, and clinical outcomes. Based on the descriptors, outcomes and a >2% threshold risk of malignancy, a follow-up algorithm was developed to decrease additional imaging without affecting cancer detection. Results: A total of 331/3800 (8.7%) patients (178 men, 153 women; mean age: 66 ± 7 years) undergoing LCS had lung findings that were attributed to infection or inflammation. These abnormalities were reported as potentially significant findings using the "S" modifier in 149/331 (45.0%) and as the "dominant nodule" determining the Lung-RADS category in 96/331 (29.0%). Abnormalities were multiple or multifocal in 260/331 (78.5%). Common descriptors were ground-glass (155/331; 46.8%), tree-in-bud (56/331; 16.9%), consolidation (41/331; 12.4%), and clustered (67/331; 20.2%) opacities. A follow-up chest CT outside of screening was performed within 12 months or less in 264/331 (79.8%) and within 6 months or less in 286/331 (56.2%). A total of 260/331 (78.5%) opacities resolved on follow-up imaging. Two malignancies (2/331; 0.60%) were associated with these abnormalities, and both had consolidations. Theoretical adoption of a proposed management algorithm for suspected infectious and inflammatory findings reduced unnecessary follow-up imaging by 82.6% without missing a single malignancy. Conclusions: Presumed acute infectious or inflammatory lung abnormalities are frequently encountered in the setting of LCS. These opacities are commonly multifocal and resolve on follow-up. Less than 1% are associated with malignancy. Clinical impact: Adoption of a conservative management algorithm can standardize recommendations and reduce unnecessary imaging without increasing the risk of missing a malignancy.
Collapse
|
26
|
Schoenmaekers JJAO, Paats MS, Dingemans AMC, Hendriks LEL. Central nervous system metastases and oligoprogression during treatment with tyrosine kinase inhibitors in oncogene-addicted non-small cell lung cancer: how to treat and when? Transl Lung Cancer Res 2020; 9:2599-2617. [PMID: 33489821 PMCID: PMC7815343 DOI: 10.21037/tlcr-20-459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Up to 70% of non-small cell lung cancer (NSCLC) patients develop central nervous system (CNS) metastases during the course of their disease, especially those with oncogenic drivers treated with a first-generation tyrosine kinase inhibitor (TKI), because of the relatively poor CNS penetration. CNS metastases are associated with a negative impact on quality of life and survival. As, with the introduction of newer generation TKIs, the survival rates are increasing in this particular population, treatment and/or prevention of CNS metastases becomes even more relevant and the TKI with the best CNS efficacy should be selected. Unfortunately, CNS efficacy data in clinical trials are not fully comparable. Furthermore, oligoprogression to the brain without extracranial progression regularly occurs in the oncogenic driver population and both local therapy and switch of systemic therapy are possible treatment options. However, the best order of systemic and local therapy is still not precisely known. In this narrative review, we will summarize incidence and treatment of CNS metastases in oncogene driven NSCLC, including the optimal treatment of CNS oligometastatic disease (synchronous as well as oligoprogressive).
Collapse
Affiliation(s)
- Janna Josephus Anna Oda Schoenmaekers
- Department of Pulmonary Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands;,Department of Pulmonary Diseases GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marthe Sentijna Paats
- Department of Pulmonary Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Anne-Marie Clasina Dingemans
- Department of Pulmonary Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands;,Department of Pulmonary Diseases GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands;,Department of Pulmonary Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Lizza Elisabeth Lucia Hendriks
- Department of Pulmonary Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands;,Department of Pulmonary Diseases GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
27
|
Garrana SH, Dagogo-Jack I, Cobb R, Kuo AH, Mendoza DP, Zhang EW, Heeger A, Sequist LV, Digumarthy SR. Clinical and Imaging Features of Non-Small-Cell Lung Cancer in Young Patients. Clin Lung Cancer 2020; 22:23-31. [PMID: 33189594 DOI: 10.1016/j.cllc.2020.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) in young adult patients is rare, with scarce data available in patients aged < 40 years and even less in those aged < 35 years. Our goal was to determine the presenting symptoms, clinicopathologic characteristics, and imaging features of young patients with NSCLC at time of diagnosis and compare them to those of older adults. PATIENTS AND METHODS We retrospectively analyzed the medical records and imaging of young patients (≤ 40 years old) with NSCLC treated at our institution between 1998 and 2018. Patients < 35 years old were compared to those between 35 and 40 years old. Characteristics of patients ≤ 40 years old were compared to older patients (> 40 years) from publicly available data sets. RESULTS We identified 166 young patients with NSCLC (median age, 36.6 years; range, 18-40 years). Most presented with nonspecific respiratory symptoms and were diagnosed with pneumonia (84/136, 62%). Compared to patients < 35 years old, patients 35-40 years old were more likely to have malignancy detected incidentally (15% vs. 5%, P = .04). Patients < 35 years old were more likely to have central tumors (55% vs. 33%, P = .02) and to have bone (38% vs. 19%, P = .007) and lung (39% vs. 24%, P = .03) metastases. Compared to older patients (> 40 years), young patients were more likely to be never smokers (65.0% vs. 14.7%, P < .001) and to have advanced disease (88% vs. 66%, P < .001). CONCLUSION Young patients with NSCLC often present with nonspecific symptoms and have advanced disease at diagnosis, often mimicking other pathologies. Awareness of the clinical presentation and imaging features of NSCLC in young patients may help minimize delays in diagnoses.
Collapse
Affiliation(s)
- Sherief H Garrana
- Harvard Medical School, Boston, MA; Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Ibiayi Dagogo-Jack
- Harvard Medical School, Boston, MA; Department of Medicine, Cancer Center, Massachusetts General Hospital, Boston, MA
| | - Rosemary Cobb
- Harvard Medical School, Boston, MA; Department of Medicine, Cancer Center, Massachusetts General Hospital, Boston, MA
| | - Anderson H Kuo
- Harvard Medical School, Boston, MA; Division of Cardiovascular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Dexter P Mendoza
- Harvard Medical School, Boston, MA; Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Eric W Zhang
- Harvard Medical School, Boston, MA; Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Allen Heeger
- Harvard Medical School, Boston, MA; Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Lecia V Sequist
- Harvard Medical School, Boston, MA; Department of Medicine, Cancer Center, Massachusetts General Hospital, Boston, MA
| | - Subba R Digumarthy
- Harvard Medical School, Boston, MA; Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
28
|
Mendoza DP, Piotrowska Z, Lennerz JK, Digumarthy SR. Role of imaging biomarkers in mutation-driven non-small cell lung cancer. World J Clin Oncol 2020; 11:412-427. [PMID: 32821649 PMCID: PMC7407925 DOI: 10.5306/wjco.v11.i7.412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/31/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths worldwide. The treatment of non-small cell lung cancer (NSCLC), which accounts for a vast majority of lung cancers, has shifted to personalized, targeted therapy following discoveries of several targetable oncogenic mutations. Targeting of specific mutations has improved outcomes in many patients. This success has led to several target-specific agents replacing chemotherapy as first-line treatment in certain mutated NSCLC. Several researchers have reported that there may be imaging biomarkers that may be predictive of the presence of these mutations. These features, when present, have the potential in triaging patients into the most appropriate diagnostic and treatment algorithms. Distinct imaging features and patterns of metastases that have been associated with NSCLC with various targetable oncogenic mutations are presented in this review.
Collapse
Affiliation(s)
- Dexter P Mendoza
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Zofia Piotrowska
- Massachusetts General Hospital Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Subba R Digumarthy
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, United States
| |
Collapse
|
29
|
Imaging Features and Patterns of Metastasis in Non-Small Cell Lung Cancer with RET Rearrangements. Cancers (Basel) 2020; 12:cancers12030693. [PMID: 32183422 PMCID: PMC7140075 DOI: 10.3390/cancers12030693] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/05/2020] [Accepted: 03/13/2020] [Indexed: 02/08/2023] Open
Abstract
Rearranged during transfection proto-oncogene (RET) fusions represent a potentially targetable oncogenic driver in non-small cell lung cancer (NSCLC). Imaging features and metastatic patterns of advanced RET fusion-positive (RET+) NSCLC are not well established. Our goal was to compare the imaging features and patterns of metastases in RET+, ALK+ and ROS1+ NSCLC. Patients with RET+, ALK+, or ROS1+ NSCLC seen at our institution between January 2014 and December 2018 with available pre-treatment imaging were identified. The clinicopathologic features, imaging characteristics, and the distribution of metastases were reviewed and compared. We identified 215 patients with NSCLC harboring RET, ALK, or ROS1 gene fusion (RET = 32; ALK = 116; ROS1 = 67). Patients with RET+ NSCLC were older at presentation compared to ALK+ and ROS1+ patients (median age: RET = 64 years; ALK = 51 years, p < 0.001; ROS = 54 years, p = 0.042) and had a higher frequency of neuroendocrine histology (RET = 12%; ALK = 2%, p = 0.025; ROS1 = 0%, p = 0.010). Primary tumors in RET+ patients were more likely to be peripheral (RET = 69%; ALK = 47%, p = 0.029; ROS1 = 36%, p = 0.003), whereas lobar location, size, and density were comparable across the three groups. RET+ NSCLC was associated with a higher frequency of brain metastases at diagnosis compared to ROS1+ NSCLC (RET = 32%, ROS1 = 10%; p = 0.039. Metastatic patterns were otherwise similar across the three molecular subgroups, with high incidences of lymphangitic carcinomatosis, pleural metastases, and sclerotic bone metastases. RET+ NSCLC shares several distinct radiologic features and metastatic spread with ALK+ and ROS1+ NSCLC. These features may suggest the presence of RET fusions and help identify patients who may benefit from further molecular genotyping.
Collapse
|