101
|
Kanemura H, Takeda M, Nakagawa K. Simultaneous targeting of MET overexpression in EGFR mutation-positive non-small cell lung cancer can increase the benefit of EGFR-TKI therapy? Transl Lung Cancer Res 2020; 9:1617-1622. [PMID: 32953535 PMCID: PMC7481636 DOI: 10.21037/tlcr-20-707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Hiroaki Kanemura
- Department of Medical Oncology, Kindai University, Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University, Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University, Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
102
|
Ding N, You A, Tian W, Gu L, Deng D. Chidamide increases the sensitivity of Non-small Cell Lung Cancer to Crizotinib by decreasing c- MET mRNA methylation. Int J Biol Sci 2020; 16:2595-2611. [PMID: 32792859 PMCID: PMC7415423 DOI: 10.7150/ijbs.45886] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction: Crizotinib is a kinase inhibitor targeting c-MET/ALK/ROS1 used as the first-line chemical for the treatment of non-small cell lung cancer (NSCLC) with ALK mutations. Although c-MET is frequently overexpressed in 35-72% of NSCLC, most NSCLCs are primarily resistant to crizotinib treatment. Method: A set of NSCLC cell lines were used to test the effect of chidamide on the primary crizotinib resistance in vitro and in vivo. Relationships between the synergistic effect of chidamide and c-MET expression and RNA methylation were systemically studied with a battery of molecular biological assays. Results: We found for the first time that chidamide could sensitize the effect of crizotinib in a set of ALK mutation-free NSCLC cell lines, especially those with high levels of c-MET expression. Notably, chidamide could not increase the sensitivity of NSCLC cells to crizotinib cultured in serum-free medium without hepatocyte growth factor (HGF; a c-MET ligand). In contrast, the addition of HGF into the serum-/HGF-free medium could restore the synergistic effect of chidamide. Moreover, the synergistic effect of chidamide could also be abolished either by treatment with c-MET antibody or siRNA-knockdown of c-MET expression. While cells with low or no c-MET expression were primarily resistant to chidamide-crizotinib cotreatment, enforced c-MET overexpression could increase the sensitivity of these cells to chidamide-crizotinib cotreatment. Furthermore, chidamide could decrease c-MET expression by inhibiting mRNA N6-methyladenosine (m6A) modification through the downregulation of METTL3 and WTAP expression. Chidamide-crizotinib cotreatment significantly suppressed the activity of c-MET downstream molecules. Conclusion: Chidamide downregulated c-MET expression by decreasing its mRNA m6A methylation, subsequently increasing the crizotinib sensitivity of NSCLC cells in a c-MET-/HGF-dependent manner.
Collapse
Affiliation(s)
- Nan Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China
| | - Abin You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China
| | - Wei Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China
| |
Collapse
|
103
|
Recondo G, Che J, Jänne PA, Awad MM. Targeting MET Dysregulation in Cancer. Cancer Discov 2020; 10:922-934. [PMID: 32532746 PMCID: PMC7781009 DOI: 10.1158/2159-8290.cd-19-1446] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/31/2020] [Accepted: 04/16/2020] [Indexed: 11/16/2022]
Abstract
Aberrant MET signaling can drive tumorigenesis in several cancer types through a variety of molecular mechanisms including MET gene amplification, mutation, rearrangement, and overexpression. Improvements in biomarker discovery and testing have more recently enabled the selection of patients with MET-dependent cancers for treatment with potent, specific, and novel MET-targeting therapies. We review the known oncologic processes that activate MET, discuss therapeutic strategies for MET-dependent malignancies, and highlight emerging challenges in acquired drug resistance in these cancers. SIGNIFICANCE: Increasing evidence supports the use of MET-targeting therapies in biomarker-selected cancers that harbor molecular alterations in MET. Diverse mechanisms of resistance to MET inhibitors will require the development of novel strategies to delay and overcome drug resistance.
Collapse
Affiliation(s)
- Gonzalo Recondo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
104
|
Optimal Management of Patients with Advanced NSCLC Harboring High PD-L1 Expression and Driver Mutations. Curr Treat Options Oncol 2020; 21:60. [PMID: 32588244 DOI: 10.1007/s11864-020-00750-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Patients with stage IV or recurrent/metastatic non-small cell lung cancer (NSCLC) whose tumors harbor high PD-L1 expression and driver mutations with approved targeted treatments (EGFR, ALK, BRAFV600E, ROS1) should receive initial therapy with targeted therapy based on impressive clinical activity. PD-(L)1 inhibitors have demonstrated minimal activity in many driver mutation subsets including EGFR and ALK and appears to have more benefit in smoking-associated oncogenic drivers (KRAS, BRAF). For KRAS-driven tumors, co-mutations such as STK11/LKB1 are negative predictive markers of immunotherapy with or without chemotherapy. Therefore, driver mutations need to be evaluated before pursuing immunotherapy independent of PD-L1 expression level. Caution should be used with TKIs following or concurrent with immunotherapy owing to potentially increased toxicity. New immunotherapy combinations are needed especially for oncogene-driven tumors associated with never or light smoking history.
Collapse
|
105
|
Li Y, Dong S, Tamaskar A, Wang H, Zhao J, Ma H, Zhao Y. Proteasome Inhibitors Diminish c-Met Expression and Induce Cell Death in Non-Small Cell Lung Cancer Cells. Oncol Res 2020; 28:497-507. [PMID: 32580819 PMCID: PMC7751221 DOI: 10.3727/096504020x15929939001042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and accounts for 85% of all lung carcinomas. The hepatocyte growth factor receptor (c-Met) has been considered as a potential therapeutic target for NSCLC. Proteasome inhibition induces cell apoptosis and has been used as a novel therapeutic approach for treating diseases including NSCLC; however, the effects of different proteasome inhibitors on NSCLC have not been fully investigated. The aim of this study is to determine a precise strategy for treating NSCLC by targeting c-Met using different proteasome inhibitors. Three proteasome inhibitors, bortezomib, MG132, and ONX 0914, were used in this study. Bortezomib (50 nM) significantly reduced c-Met levels and cell viability in H1299 and H441 cells, while similar effects were observed in H460 and A549 cells when a higher concentration (∼100 nM) was used. Bortezomib decreased c-Met gene expression in H1299 and H441 cells, but it had no effect in A549 and H460 cells. MG-132 at a low concentration (0.5 μM) diminished c-Met levels in H441 cells, while neither a low nor a high concentration (∼20 μM) altered c-Met levels in A549 and H460 cells. A higher concentration of MG-132 (5 μM) was required for decreasing c-Met levels in H1299 cells. Furthermore, MG-132 induced cell death in all four cell types. Among all the four cell lines, H441 cells expressed higher levels of c-Met and appeared to be the most susceptible to MG-132. MG-132 decreased c-Met mRNA levels in both H1299 and H441 cells. ONX 0914 reduced c-Met levels in H460, H1299, and H441 cells but not in A549 cells. c-Met levels were decreased the most in H441 cells treated with ONX 0914. ONX 0914 did not alter cell viability in H441; however, it did induce cell death among H460, A549, and H1299 cells. This study reveals that different proteasome inhibitors produce varied inhibitory effects in NSCLS cell lines.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Anesthesia, the First Hospital of Jilin UniversityChangchun, JilinP.R. China
| | - Su Dong
- Department of Anesthesia, the First Hospital of Jilin UniversityChangchun, JilinP.R. China
| | - Arya Tamaskar
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OHUSA
| | - Heather Wang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OHUSA
| | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OHUSA
| | - Haichun Ma
- Department of Anesthesia, the First Hospital of Jilin UniversityChangchun, JilinP.R. China
| | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OHUSA
| |
Collapse
|
106
|
Bylicki O, Paleiron N, Assié JB, Chouaïd C. Targeting the MET-Signaling Pathway in Non-Small-Cell Lung Cancer: Evidence to Date. Onco Targets Ther 2020; 13:5691-5706. [PMID: 32606781 PMCID: PMC7306460 DOI: 10.2147/ott.s219959] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/30/2020] [Indexed: 12/19/2022] Open
Abstract
The c-MET proto-oncogene (MET) plays an important role in lung oncogenesis, affecting cancer-cell survival, growth and invasiveness. The MET receptor in non-small-cell lung cancer (NSCLC) is a potential therapeutic target. The development of high-output next-generation sequencing techniques has enabled better identification of anomalies in the MET pathway, like the MET exon-14 (METex14) mutation. Moreover, analyses of epidermal growth factor-receptor (EGFR) and mechanisms of resistance to tyrosine-kinase inhibitors (TKIs) demonstrated the importance of MET amplification as an escape mechanism in patients with TKI-treated EGFR-mutated NSCLCs. This review summarizes the laboratory findings on MET and its anomalies, trial results on METex14 alterations and MET amplification in non-EGFR mutated NSCLCs, and acquired resistance to TKI in EGFR-mutated NSCLCs. The outcomes of the first trials with anti-MET agents on non-selected NSCLC patients or those selected for MET overexpression were disappointing. Two situations seem the most promising today for the use of anti-MET agents to treat these patients: tumors harboring METex14 and those EGFR-sensitive mutation mutated under TKI-EGFR with a MET-amplification mechanism of resistance or EGFR-resistance mutation.
Collapse
Affiliation(s)
- Olivier Bylicki
- Respiratory Disease Unit, HIA Sainte Anne, Toulon, France
- University Paris–Est Créteil (UPEC), CEpiA (Clinical Epidemiology and Ageing), EA 7376- IMRB, UPEC, Créteil, France
| | | | - Jean-Baptiste Assié
- University Paris–Est Créteil (UPEC), CEpiA (Clinical Epidemiology and Ageing), EA 7376- IMRB, UPEC, Créteil, France
- Pneumology Department, Centre Hospitalier Intercommunal De Créteil, Créteil, France
- Cordeliers Research Center, Inserm, Functional Genomics of Solid Tumors Laboratory, Sorbonne University, University of Paris, Paris, France
| | - Christos Chouaïd
- University Paris–Est Créteil (UPEC), CEpiA (Clinical Epidemiology and Ageing), EA 7376- IMRB, UPEC, Créteil, France
- Pneumology Department, Centre Hospitalier Intercommunal De Créteil, Créteil, France
| |
Collapse
|
107
|
Malik R, Mambetsariev I, Fricke J, Chawla N, Nam A, Pharaon R, Salgia R. MET receptor in oncology: From biomarker to therapeutic target. Adv Cancer Res 2020; 147:259-301. [PMID: 32593403 DOI: 10.1016/bs.acr.2020.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
First discovered in the 1984, the MET receptor tyrosine kinase (RTK) and its ligand hepatocyte growth factor or HGF (also known as scatter factor or SF) are implicated as key players in tumor cell migration, proliferation, and invasion in a variety of cancers. This pathway also plays a key role during embryogenesis in the development of muscular and nervous structures. High expression of the MET receptor has been shown to correlate with poor prognosis and resistance to therapy. MET exon 14 splicing variants, initially identified by us in lung cancer, is actionable through various tyrosine kinase inhibitors (TKIs). For this reason, this pathway is of interest as a therapeutic target. In this chapter we will be discussing the history of MET, the genetics of this RTK, and give some background on the receptor biology. Furthermore, we will discuss directed therapeutics, mechanisms of resistance, and the future of MET as a therapeutic target.
Collapse
Affiliation(s)
- Raeva Malik
- George Washington University Hospital, Washington, DC, United States
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | - Neal Chawla
- Department of Medicine, Advocate Illinois Masonic Medical Center, Chicago, IL, United States
| | - Arin Nam
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | - Rebecca Pharaon
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States.
| |
Collapse
|
108
|
Therapeutic Efficacy of ABN401, a Highly Potent and Selective MET Inhibitor, Based on Diagnostic Biomarker Test in MET-Addicted Cancer. Cancers (Basel) 2020; 12:cancers12061575. [PMID: 32549194 PMCID: PMC7352216 DOI: 10.3390/cancers12061575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 11/24/2022] Open
Abstract
The receptor tyrosine kinase c-MET regulates processes essential for tissue remodeling and mammalian development. The dysregulation of c-MET signaling plays a role in tumorigenesis. The aberrant activation of c-MET, such as that caused by gene amplification or mutations, is associated with many cancers. c-MET is therefore an attractive therapeutic target, and inhibitors are being tested in clinical trials. However, inappropriate patient selection criteria, such as low amplification or expression level cut-off values, have led to the failure of clinical trials. To include patients who respond to MET inhibitors, the selection criteria must include MET oncogenic addiction. Here, the efficacy of ABN401, a MET inhibitor, was investigated using histopathologic and genetic analyses in MET-addicted cancer cell lines and xenograft models. ABN401 was highly selective for 571 kinases, and it inhibited c-MET activity and its downstream signaling pathway. We performed pharmacokinetic profiling of ABN401 and defined the dose and treatment duration of ABN401 required to inhibit c-MET phosphorylation in xenograft models. The results show that the efficacy of ABN401 is associated with MET status and they highlight the importance of determining the cut-off values. The results suggest that clinical trials need to establish the characteristics of each sample and their correlations with the efficacy of MET inhibitors.
Collapse
|
109
|
Rajurkar S, Mambetsariev I, Pharaon R, Leach B, Tan T, Kulkarni P, Salgia R. Non-Small Cell Lung Cancer from Genomics to Therapeutics: A Framework for Community Practice Integration to Arrive at Personalized Therapy Strategies. J Clin Med 2020; 9:E1870. [PMID: 32549358 PMCID: PMC7356243 DOI: 10.3390/jcm9061870] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a heterogeneous disease, and therapeutic management has advanced with the identification of various key oncogenic mutations that promote lung cancer tumorigenesis. Subsequent studies have developed targeted therapies against these oncogenes in the hope of personalizing therapy based on the molecular genomics of the tumor. This review presents approved treatments against actionable mutations in NSCLC as well as promising targets and therapies. We also discuss the current status of molecular testing practices in community oncology sites that would help to direct oncologists in lung cancer decision-making. We propose a collaborative framework between community practice and academic sites that can help improve the utilization of personalized strategies in the community, through incorporation of increased testing rates, virtual molecular tumor boards, vendor-based oncology clinical pathways, and an academic-type singular electronic health record system.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (S.R.); (I.M.); (R.P.); (B.L.); (T.T.); (P.K.)
| |
Collapse
|
110
|
Guo R, Luo J, Chang J, Rekhtman N, Arcila M, Drilon A. MET-dependent solid tumours - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol 2020; 17:569-587. [PMID: 32514147 DOI: 10.1038/s41571-020-0377-z] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Attempts to develop MET-targeted therapies have historically focused on MET-expressing cancers, with limited success. Thus, MET expression in the absence of a genomic marker of MET dependence is a poor predictor of benefit from MET-targeted therapy. However, owing to the development of more sensitive methods of detecting genomic alterations, high-level MET amplification and activating MET mutations or fusions are all now known to be drivers of oncogenesis. MET mutations include those affecting the kinase or extracellular domains and those that result in exon 14 skipping. The activity of MET tyrosine kinase inhibitors varies by MET alteration category. The likelihood of benefit from MET-targeted therapies increases with increasing levels of MET amplification, although no consensus exists on the optimal diagnostic cut-off point for MET copy number gains identified using fluorescence in situ hybridization and, in particular, next-generation sequencing. Several agents targeting exon 14 skipping alterations are currently in clinical development, with promising data available from early-phase trials. By contrast, the therapeutic implications of MET fusions remain underexplored. Here we summarize and evaluate the utility of various diagnostic techniques and the roles of different classes of MET-targeted therapies in cancers with MET amplification, mutation and fusion, and MET overexpression.
Collapse
Affiliation(s)
- Robin Guo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jia Luo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
111
|
Wang Y, Tian P, Xia L, Li L, Han R, Zhu M, Lizaso A, Qin T, Li M, Yu B, Mao X, Han-Zhang H, He Y. The clinical efficacy of combinatorial therapy of EGFR-TKI and crizotinib in overcoming MET amplification-mediated resistance from prior EGFR-TKI therapy. Lung Cancer 2020; 146:165-173. [PMID: 32540560 DOI: 10.1016/j.lungcan.2020.06.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND MET amplification is one of the EGFR-independent mechanisms of EGFR tyrosine kinase inhibitor (TKI) resistance. Combinatorial therapy of EGFR-TKI and crizotinib has been explored as a strategy to overcome resistance by simultaneously targeting both EGFR and MET pathways; however, no consensus still exists on the optimal combination regimen with the most benefit. METHODS Retrospective analysis was performed on the clinical and sequencing data obtained from eleven patients with lung adenocarcinoma who acquired MET amplification at progression from prior EGFR-TKI therapy and received a combination of EGFR-TKI and crizotinib. RESULTS Acquired MET amplification was detected in four and seven patients who progressed from first-line gefitinib and second-line osimertinib, respectively. Six and five patients received a combination of either first-generation (gefitinib, erlotinib, or icotinib) or third-generation (osimertinib) EGFR-TKI and crizotinib, respectively. Nine patients achieved partial response, resulting in an overall response rate of 81.8 %. The median progression-free survival of the cohort was 5.8 months. Moreover, analysis of acquired resistance mechanisms from nine patients identified EGFR T790 M from three patients who progressed from first-generation EGFR-TKI and crizotinib, while EGFR T790 M/trans-C797S and L718Q, EGFR G724S, and CCDC6-RET fusion were detected from one patient each who progressed from osimertinib and crizotinib regimen. Loss of MET amplification was also observed in a majority of the patients at progression from the combination therapy. CONCLUSIONS Our study provides clinical evidence of the efficacy of combinatorial regimen with either first- or third-generation EGFR-TKI and crizotinib after the emergence of MET amplification-mediated EGFR-TKI resistance in patients with EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Yubo Wang
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, 400042 China
| | - Panwen Tian
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Lei Xia
- Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Li Li
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, 400042 China
| | - Rui Han
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, 400042 China
| | - Mengxiao Zhu
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, 400042 China
| | | | - Tian Qin
- Burning Rock Biotech, Guangzhou, 510300 China
| | - Min Li
- Burning Rock Biotech, Guangzhou, 510300 China
| | - Bing Yu
- Burning Rock Biotech, Guangzhou, 510300 China
| | - Xinru Mao
- Burning Rock Biotech, Guangzhou, 510300 China
| | | | - Yong He
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, 400042 China.
| |
Collapse
|
112
|
Wu S, Li G, Zhao X, Xiang J, Lizaso A, Ye J, Shi C, Chen L. High-level gain of mesenchymal-epithelial transition factor (MET) copy number using next-generation sequencing as a predictive biomarker for MET inhibitor efficacy. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:685. [PMID: 32617305 PMCID: PMC7327325 DOI: 10.21037/atm-20-2741] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background In clinical oncology, targeted next-generation sequencing (NGS) has become an integral part of the routine molecular diagnostics repertoire. However, a consensus is yet to be agreed on the optimal mesenchymal-epithelial transition factor (MET) copy number (CN) cut-off value based on NGS data that could predict the MET-amplified non-small cell lung cancer (NSCLC) patients who could benefit from MET tyrosine kinase inhibitor (TKI) therapy. In this study, we aimed to identify the criteria to define MET amplification derived from NGS data. Methods Sequencing data from matched plasma and tissue samples from 40 MET-amplified NSCLC patients were used to derive a normalization method, referred to as adjusted copy number (adCN). Clinical outcomes from an additional 18 MET TKI-treated NSCLC patients with solely MET-amplified cancers were analyzed to validate the adCN cut-offs. Results AdCN, calculated as the absolute CN generated from NGS relative to the maximum mutant allele fraction (maxMAF) per sample, was demonstrated to have a high correlation with MET CN in tissue and plasma samples (R2=0.73). Using a cut-off value of 5.5 and 13, tertile stratification of adCN was able to distinguish patients with high-level MET amplification. The MET TKI-treated patients with adCN >13, categorized as high-level amplification, had significantly longer progression-free survival (PFS) than those with adCN <13 (P=0.009), suggesting that adCN positively correlated with the response to MET TKI. Conclusions We derived a normalization method that could reflect the relative CN and distinguish MET-amplified NSCLC patients with high-level gene amplification who were sensitive to crizotinib, suggesting adCN could potentially serve as a predictive biomarker for MET TKI response.
Collapse
Affiliation(s)
- Shibo Wu
- Department of Respiratory Medicine, Lihuili Hospital, Ningbo Medical Center, Ningbo, China
| | - Guodong Li
- Department of Interventional Therapy, Fudan University Shanghai Cancer Center, Shanghai China
| | - Xin Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | - Junyi Ye
- Burning Rock Biotech, Guangzhou, China
| | - Chunlei Shi
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Lingxiang Chen
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
113
|
Sugano T, Yoshida M, Masuda M, Ono M, Tamura K, Kinoshita T, Tsuda H, Honda K, Gemma A, Yamada T. Prognostic impact of ACTN4 gene copy number alteration in hormone receptor-positive, HER2-negative, node-negative invasive breast carcinoma. Br J Cancer 2020; 122:1811-1817. [PMID: 32265507 PMCID: PMC7283275 DOI: 10.1038/s41416-020-0821-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 01/08/2020] [Accepted: 03/10/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Most patients with hormone receptor (HR)-positive, human epidermal growth factor receptor type 2 (HER2)-negative breast cancer can be cured by surgery and endocrine therapy, but a significant proportion suffer recurrences. Actinin-4 is associated with cancer invasion and metastasis, and its genetic alteration may be used for breast cancer prognostication. METHODS The copy number of the actinin-4 (ACTN4) gene was determined by fluorescence in situ hybridisation (FISH) in two independent cohorts totalling 597 patients (336 from Japan and 261 from the USA) with HR-positive, HER2-negative, node-negative breast cancer. RESULTS In the Japanese cohort, multivariate analysis revealed that a copy number increase (CNI) of ACTN4 was an independent factor associated with high risks of recurrence (P = 0.01; hazard ratio (HR), 2.95) and breast cancer death (P = 0.014; HR, 4.27). The prognostic significance of ACTN4 CNI was validated in the US cohort, where it was the sole prognostic factor significantly associated with high risks of recurrence (P = 0.04; HR, 2.73) and death (P = 0.016; HR, 4.01). CONCLUSIONS Copy number analysis of a single gene, ACTN4, can identify early-stage luminal breast cancer patients with a distinct outcome. Such high-risk patients may benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Teppei Sugano
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo, 104-0045, Japan.
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8602, Japan.
| | - Masayuki Yoshida
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Mari Masuda
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Makiko Ono
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Kenji Tamura
- Departments of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Takayuki Kinoshita
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, Saitama, 359-8513, Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Tesshi Yamada
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| |
Collapse
|
114
|
Schuler M, Berardi R, Lim WT, de Jonge M, Bauer TM, Azaro A, Gottfried M, Han JY, Lee DH, Wollner M, Hong DS, Vogel A, Delmonte A, Akimov M, Ghebremariam S, Cui X, Nwana N, Giovannini M, Kim TM. Molecular correlates of response to capmatinib in advanced non-small-cell lung cancer: clinical and biomarker results from a phase I trial. Ann Oncol 2020; 31:789-797. [PMID: 32240796 PMCID: PMC9720758 DOI: 10.1016/j.annonc.2020.03.293] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Dysregulation of receptor tyrosine kinase MET by various mechanisms occurs in 3%-4% of non-small-cell lung cancer (NSCLC) and is associated with unfavorable prognosis. While MET is a validated drug target in lung cancer, the best biomarker strategy for the enrichment of a susceptible patient population still remains to be defined. Towards this end we analyze here primary data from a phase I dose expansion study of the MET inhibitor capmatinib in patients with advanced MET-dysregulated NSCLC. PATIENTS AND METHODS Eligible patients [≥18 years; Eastern Cooperative Oncology Group (ECOG) performance status ≤2] with MET-dysregulated advanced NSCLC, defined as either (i) MET status by immunohistochemistry (MET IHC) 2+ or 3+ or H-score ≥150, or MET/centromere ratio ≥2.0 or gene copy number (GCN) ≥5, or (ii) epidermal growth factor receptor wild-type (EGFRwt) and centrally assessed MET IHC 3+, received capmatinib at the recommended dose of 400 mg (tablets) or 600 mg (capsules) b.i.d. The primary objective was to determine safety and tolerability; the key secondary objective was to explore antitumor activity. The exploratory end point was the correlation of clinical activity with different biomarker formats. RESULTS Of 55 patients with advanced MET-dysregulated NSCLC, 40/55 (73%) had received two or more prior systemic therapies. All patients discontinued treatment, primarily due to disease progression (69.1%). The median treatment duration was 10.4 weeks. The overall response rate per RECIST was 20% (95% confidence interval, 10.4-33.0). In patients with MET GCN ≥6 (n = 15), the overall response rate by both the investigator and central assessments was 47%. The median progression-free survival per investigator for patients with MET GCN ≥6 was 9.3 months (95% confidence interval, 3.8-11.9). Tumor responses were observed in all four patients with METex14. The most common toxicities were nausea (42%), peripheral edema (33%), and vomiting (31%). CONCLUSIONS MET GCN ≥6 and/or METex14 are suited to predict clinical activity of capmatinib in patients with NSCLC (NCT01324479).
Collapse
Affiliation(s)
- M Schuler
- Department of Medical Oncology, West German Cancer Center, University Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| | - R Berardi
- Clinica Oncologica, Università Politecnica delle Marche-Ospedali Riuniti, Ancona, Italy
| | - W-T Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - M de Jonge
- Medical Oncology, Erasmus MC Cancer Center, Rotterdam, The Netherlands
| | - T M Bauer
- Drug Development Unit, Sarah Cannon Research Institute, and Tennessee Oncology, PLCC, Nashville, USA
| | - A Azaro
- Medical Oncology, Molecular Therapeutics Research Unit, Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Pharmacology Department, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - M Gottfried
- Department of Oncology, Oncology Institute of Meir Medical Center, Tel-Aviv, Israel
| | - J-Y Han
- Center for Lung Cancer, National Cancer Center, Seoul
| | - D H Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - M Wollner
- Thoracic Service Oncology Department, Rambam Health Care Campus, Haifa, Israel
| | - D S Hong
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, USA
| | - A Vogel
- Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Hannover, Germany
| | - A Delmonte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - M Akimov
- Oncology Global Development, Novartis Pharma AG, Basel, Switzerland
| | | | - X Cui
- Novartis Institutes for Biomedical Research
| | | | - M Giovannini
- Oncology Global Development, Novartis Pharmaceuticals Corporation, East Hanover, USA
| | - T M Kim
- Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
115
|
Overbeck TR, Cron DA, Schmitz K, Rittmeyer A, Körber W, Hugo S, Schnalke J, Lukat L, Hugo T, Hinterthaner M, Reuter-Jessen K, Rosenthal T, Moecks J, Bleckmann A, Schildhaus HU. Top-level MET gene copy number gain defines a subtype of poorly differentiated pulmonary adenocarcinomas with poor prognosis. Transl Lung Cancer Res 2020; 9:603-616. [PMID: 32676323 PMCID: PMC7354108 DOI: 10.21037/tlcr-19-339] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background MET amplifications occur in human tumors, including non-small cell lung cancer (NSCLC). MET inhibitors have demonstrated some clinical activity in MET amplified NSCLC, presumably with a gene dose effect. However, the definition of MET positivity or MET amplification as a potential oncogenic driver is still under debate. In this study, we aimed to establish the molecular subgroup of NSCLC with the highest unequivocal MET amplification level and to describe the prevalence, and histologic and clinical phenotype of this subgroup. Methods A total of 373 unselected patients with NSCLC were consecutively tested for MET gene copy number (GCN) by FISH. Mean GCN, MET/CEN7 ratio and other FISH parameters were identified and correlated with morphological and molecular pathological characteristics of the tumors as well as with clinical data. Results Based on the variability of obtained data a top-level category of MET amplification was newly defined (>90th percentile of average GCN; ≥10 MET gene copies per tumor cell). This criterion was fulfilled in 2% of analyzed tumors. These tumors were exclusively poorly differentiated adenocarcinomas with a predominant solid subtype and pleomorphic features. Rarely, co-alterations were detected (KRAS mutation or MET exon 14 skipping mutation). In this top-level group, there were no EGFR mutations or ALK or ROS1 alterations. The most important clinical feature was a significantly shortened overall survival (HR 3.61; median OS 8.2 vs. 23.6 months). Worse prognosis did not depend on initial stage or treatment. Conclusions The newly defined top-level category of MET amplification in NSCLC defines a specific subgroup of pulmonary adenocarcinoma with adverse prognosis and characteristic morphological features. Lower levels of MET gene copy number seem to have probably no specific value as a prognostic or predictive biomarker.
Collapse
Affiliation(s)
- Tobias Raphael Overbeck
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany.,Lungentumorzentrum Universität Göttingen and Göttingen Comprehensive Cancer Center (G-CCC), Göttingen, Germany
| | - Dana Alina Cron
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Katja Schmitz
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany.,Innpath GmbH, Innsbruck, Austria
| | - Achim Rittmeyer
- Lungentumorzentrum Universität Göttingen and Göttingen Comprehensive Cancer Center (G-CCC), Göttingen, Germany.,Lungenfachklinik Immenhausen, Immenhausen, Germany
| | - Wolfgang Körber
- Lungentumorzentrum Universität Göttingen and Göttingen Comprehensive Cancer Center (G-CCC), Göttingen, Germany.,Abteilung Pneumologie, Beatmungsmedizin und Schlaflabor, Evangelisches Krankenhaus Weende, Göttingen, Germany
| | - Sara Hugo
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Juliane Schnalke
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Laura Lukat
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Tabea Hugo
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Marc Hinterthaner
- Lungentumorzentrum Universität Göttingen and Göttingen Comprehensive Cancer Center (G-CCC), Göttingen, Germany.,Department of Heart, Thoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Kirsten Reuter-Jessen
- Lungentumorzentrum Universität Göttingen and Göttingen Comprehensive Cancer Center (G-CCC), Göttingen, Germany.,Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Tessa Rosenthal
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Annalen Bleckmann
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany.,Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Hans-Ulrich Schildhaus
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany.,Institute of Pathology, University Hospital Essen, Essen, Germany.,West German Cancer Center (WTZ), Essen, Germany
| |
Collapse
|
116
|
Wu YL, Cheng Y, Zhou J, Lu S, Zhang Y, Zhao J, Kim DW, Soo RA, Kim SW, Pan H, Chen YM, Chian CF, Liu X, Tan DSW, Bruns R, Straub J, Johne A, Scheele J, Park K, Yang JCH. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): an open-label, phase 1b/2, multicentre, randomised trial. THE LANCET RESPIRATORY MEDICINE 2020; 8:1132-1143. [PMID: 32479794 DOI: 10.1016/s2213-2600(20)30154-5] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND We evaluated the efficacy and safety of tepotinib, a potent and highly selective oral MET inhibitor, plus gefitinib in patients with epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) with MET overexpression (immunohistochemistry [IHC]2+ or IHC3+) or MET amplification having acquired resistance to EGFR inhibition. METHODS In this open-label, phase 1b/2, multicentre, randomised trial (the INSIGHT study), we enrolled adult patients (≥18 years) with advanced or metastatic NSCLC, and Eastern Cooperative Oncology Group performance status of 0 or 1, from academic medical centres and community clinics in six Asian countries. In phase 1b, patients received oral tepotinib 300 mg or 500 mg plus gefitinib 250 mg once daily. In phase 2, patients with EGFR-mutant, T790M-negative NSCLC MET overexpression or MET amplification were randomly assigned (initially in a 1:1 ratio and then 2:1 following a protocol amendment) to tepotinib plus gefitinib at the recommended phase 2 dose or to standard platinum doublet chemotherapy. Randomisation was done centrally via an interactive voice-response system. The primary endpoint was investigator-assessed progression-free survival (PFS). Secondary endpoints included overall survival (OS) and safety. Subgroup analyses were preplanned in patients with high MET overexpression (IHC3+) or MET amplification (mean gene copy number ≥5 or MET to centromere of chromosome 7 ratio ≥2). Efficacy and patient characteristics were assessed on an intention-to-treat basis and safety was assessed for all patients who received at least one dose of study medication. Low recruitment led to early termination of phase 2, so all analyses are considered to be exploratory. This study is registered with ClinicalTrials.gov, NCT01982955, and the European Union Drug Regulating Authorities Clinical Trials Database, Eudra-CT 2016-001604-28. FINDINGS From Dec 23, 2013, to May 25, 2017, 18 patients were enrolled in phase 1b (n=6 in the 300 mg tepotinib group; n=12 in the 500 mg tepotinib group) and 55 patients in phase 2 (n=31 in the tepotinib plus gefitinib group; n=24 in the chemotherapy group). No dose-limiting toxicities were observed in phase 1b, so tepotinib 500 mg was used as the recommended phase 2 dose. In phase 2, survival outcomes were similar between groups: median PFS was 4·9 months in the tepotinib plus gefitinib group (90% CI 3·9-6·9) versus 4·4 months in the chemotherapy group (90% CI 4·2-6·8; hazard ratio [HR] 0·67, 90% CI 0·35-1·28). Median OS was 17·3 months in the tepotinib plus gefitinib group (12·1-37·3) versus 18·7 months in the chemotherapy group (15·9-20·7; HR 0·69, 0·34-1·41). PFS and OS were longer with tepotinib plus gefitinib than with chemotherapy in patients with high (IHC3+) MET overexpression (n=34; median PFS 8·3 months [4·1-16·6] vs 4·4 months [4·1-6·8]; HR 0·35, 0·17-0·74; median OS 37·3 months [90% CI 24·2-37·3] vs 17·9 months [12·0-20·7]; HR 0·33, 0·14-0·76) or MET amplification (n=19; median PFS 16·6 months [8·3-not estimable] vs 4·2 months [1·4-7·0]; HR 0·13, 0·04-0·43; median OS 37·3 months [90% CI not estimable] vs 13·1 months [3·25-not estimable]; HR 0·08, 0·01-0·51). The most frequent treatment-related grade 3 or worse adverse events were increased amylase (5 [16%] of 31 patients) and lipase (4 [13%]) concentrations in the tepotinib plus gefitinib group and anaemia (7 [30%] of 23 patients) and decreased neutrophil count (3 [13%]) in the chemotherapy group. INTERPRETATION Despite early study termination, in a preplanned subgroup analysis, our findings suggest improved anti activity for tepotinib plus gefitinib compared with standard chemotherapy in patients with EGFR-mutant NSCLC and MET amplification, warranting further exploration. FUNDING Merck KGaA.
Collapse
Affiliation(s)
- Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Ying Cheng
- Jilin Province Cancer Hospital, Changchun, China
| | - Jianying Zhou
- The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Shun Lu
- Shanghai Chest Hospital, Shanghai, China
| | | | - Jun Zhao
- Beijing Cancer Hospital, Beijing, China
| | - Dong-Wan Kim
- Seoul National University Hospital, Seoul, South Korea
| | | | | | - Hongming Pan
- Sir Run Run Shaw Hospital, Zhejiang Medical University, Zhejiang, China
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veteran General Hospital and School of Medicine, Taipei, Taiwan
| | - Chih-Feng Chian
- Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Xiaoqing Liu
- Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | | | - Rolf Bruns
- Biostatistics, Merck KGaA, Darmstadt, Germany
| | - Josef Straub
- Translational Medicine, Department of Clinical Biomarkers and Companion Diagnostics, Merck KGaA, Darmstadt, Germany
| | - Andreas Johne
- Global Clinical Development, Merck KGaA, Darmstadt, Germany
| | - Jürgen Scheele
- Global Clinical Development, Merck KGaA, Darmstadt, Germany
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | |
Collapse
|
117
|
Hu H, Chen F, Dong Y, Liu Y, Gong P. Discovery of novel dual c-Met/HDAC inhibitors as a promising strategy for cancer therapy. Bioorg Chem 2020; 101:103970. [PMID: 32480172 DOI: 10.1016/j.bioorg.2020.103970] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/06/2023]
Abstract
Owing to the low efficacy and acquired resistance in clinical trials of c-Met inhibitors, based on the synergistic effects between c-Met and HDAC, novel c-Met and HDAC dual inhibitors were designed and synthesized. We introduced 2-pyrrolidinone to form the 5-atoms linker for c-Met inhibitor and hydroxamic acid as a zinc binding motif for HDAC inhibitor. The highly active dual inhibitor 15f showed excellent and balanced activity against both c-Met (IC50 = 12.50 nM) and HDAC1 (IC50 = 26.97 nM). In those tested tumor cell lines, 15f exhibits efficient antiproliferative activity with greater potency than Vorinostat (SAHA) and Cabozantinib (XL184). However, by comparing with an equimolar mixture of SAHA and Foretinib, we did not observe the compounds showed clearly synergistic antiproliferative effect. Nevertheless, compound 15f was found to induce apoptosis and cause cell cycle arrest in G2/M phase. This proof-of-concept study provides an efficient strategy for discovery of multitarget antitumor drugs.
Collapse
Affiliation(s)
- Hao Hu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Fei Chen
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yuhong Dong
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yajing Liu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| | - Ping Gong
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| |
Collapse
|
118
|
Design, synthesis and biological evaluation of novel N-sulfonylamidine-based derivatives as c-Met inhibitors via Cu-catalyzed three-component reaction. Eur J Med Chem 2020; 200:112470. [PMID: 32505087 DOI: 10.1016/j.ejmech.2020.112470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022]
Abstract
In our continuing efforts to develop novel c-Met inhibitors as potential anticancer candidates, a series of new N-sulfonylamidine derivatives were designed, synthesized via Cu-catalyzed multicomponent reaction (MCR) as the key step, and evaluated for their in vitro biological activities against c-Met kinase and four cancer cell lines (A549, HT-29, MKN-45 and MDA-MB-231). Most of the target compounds showed moderate to significant potency at both the enzyme-based and cell-based assay and possessed selectivity for A549 and HT-29 cancer cell lines. The preliminary SAR studies demonstrated that compound 26af (c-Met IC50 = 2.89 nM) was the most promising compound compared with the positive foretinib, which exhibited the remarkable antiproliferative activities, with IC50 values ranging from 0.28 to 0.72 μM. Mechanistic studies of 26af showed the anticancer activity was closely related to the blocking phosphorylation of c-Met, leading to cell cycle arresting at G2/M phase and apoptosis of A549 cells by a concentration-dependent manner. The promising compound 26af was further identified as a relatively selective inhibitor of c-Met kinase, which also possessed an acceptable safety profile and favorable pharmacokinetic properties in BALB/c mouse. The favorable drug-likeness of 26af suggested that N-sulfonylamidines may be used as a promising scaffold for antitumor drug development. Additionally, the docking study and molecular dynamics simulations of 26af revealed a common mode of interaction with the binding site of c-Met. These positive results indicated that compound 26af is a potential anti-cancer candidate for clinical trials, and deserves further development as a selective c-Met inhibitor.
Collapse
|
119
|
Jénot L, Rouquette I, Mazieres J. [Management of a patient with a double EGFR and MET anomaly by combined treatment]. Rev Mal Respir 2020; 37:488-491. [PMID: 32430158 DOI: 10.1016/j.rmr.2020.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 03/30/2020] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Lung cancer displays molecular anomalies for which targeted therapies are the standard first line treatment. The EGFR mutation is present in 10% of cases of non-small cell lung cancer in Caucasians. MET amplification associated with an exon 19 EGFR mutation has been identified though it is usually regarded as a mechanism of resistance. CASE REPORT We report the case of a 74-year-old never-smoking woman who was diagnosed with stage IV bronchial adenocarcinoma showing both EGFR mutation and MET amplification. Initial treatment with gefitinib did not control the disease. Platinum-based chemotherapy with pemetrexed maintenance allowed a temporary response. Treatment with durvalumab for 27 months was associated with disease stability. Single agent crizotinib was associated with a slight response followed by progression. The concomitant introduction of crizotinib and gefitinib led to a spectacular and durable response with no safety issues. CONCLUSIONS This case highlights the efficacy of concomitant treatment in a patient with two oncogenic drivers.
Collapse
Affiliation(s)
- L Jénot
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, France
| | - I Rouquette
- Service d'anatomopathologie, institut universitaire du cancer de Toulouse, CHU Toulouse, France
| | - J Mazieres
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, France.
| |
Collapse
|
120
|
Nan X, Li HJ, Fang SB, Li QY, Wu YC. Structure-based discovery of novel 4-(2-fluorophenoxy)quinoline derivatives as c-Met inhibitors using isocyanide-involved multicomponent reactions. Eur J Med Chem 2020; 193:112241. [DOI: 10.1016/j.ejmech.2020.112241] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/01/2020] [Accepted: 03/13/2020] [Indexed: 11/29/2022]
|
121
|
Hamid AB, Petreaca RC. Secondary Resistant Mutations to Small Molecule Inhibitors in Cancer Cells. Cancers (Basel) 2020; 12:cancers12040927. [PMID: 32283832 PMCID: PMC7226513 DOI: 10.3390/cancers12040927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Secondary resistant mutations in cancer cells arise in response to certain small molecule inhibitors. These mutations inevitably cause recurrence and often progression to a more aggressive form. Resistant mutations may manifest in various forms. For example, some mutations decrease or abrogate the affinity of the drug for the protein. Others restore the function of the enzyme even in the presence of the inhibitor. In some cases, resistance is acquired through activation of a parallel pathway which bypasses the function of the drug targeted pathway. The Catalogue of Somatic Mutations in Cancer (COSMIC) produced a compendium of resistant mutations to small molecule inhibitors reported in the literature. Here, we build on these data and provide a comprehensive review of resistant mutations in cancers. We also discuss mechanistic parallels of resistance.
Collapse
|
122
|
Liang H, Wang M. MET Oncogene in Non-Small Cell Lung Cancer: Mechanism of MET Dysregulation and Agents Targeting the HGF/c-Met Axis. Onco Targets Ther 2020; 13:2491-2510. [PMID: 32273721 PMCID: PMC7104217 DOI: 10.2147/ott.s231257] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/16/2020] [Indexed: 12/24/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide and has a poor prognosis. Current treatments for advanced NSCLC included traditional chemotherapy, radiotherapy, targeted therapy, and immunotherapy. The efficacy of targeted therapy relies on oncogene addiction. Mesenchymal-epithelial transition factor (MET) gene can encode unconventional receptor tyrosine kinases with pleiotropic functions, when signals are abnormally activated, it can initiate and maintain tumor transformation, promote cell proliferation, survival, tumor invasion and angiogenesis. Thus, it is a promising therapeutic target. Previous studies have shown that elevated levels of HGF and/or overexpression of c-Met are associated with poor prognosis in lung cancer. In preclinical and clinical trials, c-MET inhibitors have shown some antitumor activity in NSCLC. Although the efficacy results of MET inhibitors in Phase III clinical trials are disappointing, given the molecular heterogeneity of NSCLC, only subgroups of patients with MET gene alterations may benefit from c-MET inhibitors. The challenge for the future is to screen out the potential beneficiaries. To solve this problem, there is need for large data analysis for the detection methods and treatment effects, to establish standards that meet the MET activation status, and determine reliable thresholds to achieve effective patient stratification and clinical decision making. This article summarized the structure of the hepatocyte growth factor (HGF)/c-Met axis, the different mechanisms of MET addiction, as well as MET amplification as acquired resistance mechanism to epidermal growth factor receptor-tyrosine kinase inhibitors, the latest advances of MET inhibitors, and immuotherapy in the treatment of NSCLC with MET alterations.
Collapse
Affiliation(s)
- Hongge Liang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing100730, People’s Republic of China
| | - Mengzhao Wang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing100730, People’s Republic of China
| |
Collapse
|
123
|
Abstract
Lung cancer is a heterogeneous genomic disease. Smoking remains the primary cause. Genetic susceptibility and environmental exposures are responsible for 10% to 15% of cases. Targeted therapies improve survival in patients with tumors with oncogenic drivers. It is critical to expand our understanding of genetic alterations in non-small cell lung cancer to increase the available targeted therapies. Alterations beyond epidermal growth factor receptor (EGFR), ALK, and ROS1 exemplify lung cancer's complexity and the need for investments in precision therapy to extend patient survival and improve outcomes. This article covers genetic targets beyond EGFR, ALK and ROS1, their novel agents, challenges, and future directions.
Collapse
Affiliation(s)
- Karen L Reckamp
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| |
Collapse
|
124
|
Detection of MET Alterations Using Cell Free DNA and Circulating Tumor Cells from Cancer Patients. Cells 2020; 9:cells9020522. [PMID: 32102486 PMCID: PMC7072825 DOI: 10.3390/cells9020522] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/05/2020] [Accepted: 02/21/2020] [Indexed: 12/24/2022] Open
Abstract
MET alterations may provide a potential biomarker to evaluate patients who will benefit from treatment with MET inhibitors. Therefore, the purpose of the present study is to investigate the utility of a liquid biopsy-based strategy to assess MET alterations in cancer patients. We analyzed MET amplification in circulating free DNA (cfDNA) from 174 patients with cancer and 49 healthy controls and demonstrated the accuracy of the analysis to detect its alteration in patients. Importantly, a significant correlation between cfDNA concentration and MET copy number (CN) in cancer patients (r = 0.57, p <10−10) was determined. Furthermore, we evaluated two approaches to detect the presence of MET on circulating tumor cells (CTCs), using the CellSearch® and Parsortix systems and monitored patients under anti-EGFR treatment (n = 30) combining both cfDNA and CTCs analyses. This follow-up provides evidence for the potential of MET CN assessment when patients develop resistance to anti-EGFR therapy and a significant association between the presence of CTCs MET+ and the Overall Survival (OS) in head and neck cancer patients (P = 0.05; HR = 6.66). In conclusion, we develop specific and noninvasive assays to monitor MET status in cfDNA/CTCs and demonstrate the utility of plasma MET CN determination as a biomarker for monitoring the appearance of resistance to anti-EGFR therapy.
Collapse
|
125
|
Hara T, Kimura A, Miyazaki T, Tanaka H, Morimoto M, Nakai K, Soeda J. Cabozantinib inhibits AXL- and MET-dependent cancer cell migration induced by growth-arrest-specific 6 and hepatocyte growth factor. Biochem Biophys Rep 2020; 21:100726. [PMID: 32055714 PMCID: PMC7005370 DOI: 10.1016/j.bbrep.2020.100726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
Cabozantinib is known as an inhibitor of receptor tyrosine kinases mainly targeting AXL receptor tyrosine kinase (AXL), MET proto-oncogene-encoded receptor tyrosine kinase (MET), and vascular endothelial growth factor receptor 2. Growth arrest-specific 6 (GAS6) and hepatocyte growth factor (HGF), the natural ligands of AXL and MET, respectively, are associated with the induction of cancer cell proliferation or metastasis. Currently, it is still unclear how cabozantinib regulates cancer cell migration and invasion by inhibiting AXL and MET. This study was conducted to investigate the mechanism underlying the anti-cancer effects of cabozantinib through regulation of AXL and MET signaling. The results of Boyden chamber assays showed that cancer cell migration was induced by GAS6 and HGF in SKOV3 cells in serum-free medium. Combinatorial treatment with GAS6 and HGF exerted an additive effect on cell migration. Furthermore, we examined the role of AXL and MET signaling in cell migration. Short interfering RNA targeting AXL and MET inhibited GAS6- and HGF-induced migration, respectively. Double knockdown of AXL and MET completely suppressed cell migration induced by combination treatment with GAS6 and HGF compared to AXL or MET inhibition alone. Finally, we investigated the effects of cabozantinib on cell migration and invasion. Cabozantinib inhibited AXL and MET phosphorylation and downregulated the downstream mediators, phosphorylated SRC in the presence of both GAS6 and HGF in SKOV3 cells. The cell migration and invasion induced by combined GAS6 and HGF treatment were suppressed by cabozantinib, but not by capmatinib, a selective MET inhibitor. Our data indicate that the GAS6-AXL and HGF-MET signal pathways markedly contribute to cancer cell migration and invasion in an independent manner, suggesting that simultaneous inhibition of these two pathways contributes to the anti-cancer effects of cabozantinib.
Collapse
Affiliation(s)
- Takahito Hara
- Innovation Promotion, Shonan Research Central Office, Research, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka-Higashi, Fujisawa-shi, Kanagawa, 251-8555, Japan
| | - Akiko Kimura
- Oncology Therapeutic Area Unit for Japan & Asia, Takeda Pharmaceutical Company Limited, 4-1-1 Dosho-machi Chuo-ku Osaka-shi, Osaka, 540-8645, Japan
| | - Tohru Miyazaki
- Department of Japan Medical Affairs, Japan Oncology Business Unit, Takeda Pharmaceutical Company Limited, 2-1-1 Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8668, Japan
| | - Hiroshi Tanaka
- Axcelead Drug Discovery Partners, Inc., 2-26-1Muraoka-Higashi, Fujisawa-shi, Kanagawa, 251-0012, Japan
| | - Megumi Morimoto
- Innovation Promotion, Shonan Research Central Office, Research, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka-Higashi, Fujisawa-shi, Kanagawa, 251-8555, Japan
| | - Katsuhiko Nakai
- Oncology Therapeutic Area Unit for Japan & Asia, Takeda Pharmaceutical Company Limited, 4-1-1 Dosho-machi Chuo-ku Osaka-shi, Osaka, 540-8645, Japan
| | - Junpei Soeda
- Department of Japan Medical Affairs, Japan Oncology Business Unit, Takeda Pharmaceutical Company Limited, 2-1-1 Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8668, Japan
| |
Collapse
|
126
|
Salgueiro L, Buccitelli C, Rowald K, Somogyi K, Kandala S, Korbel JO, Sotillo R. Acquisition of chromosome instability is a mechanism to evade oncogene addiction. EMBO Mol Med 2020; 12:e10941. [PMID: 32030896 PMCID: PMC7059010 DOI: 10.15252/emmm.201910941] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 01/04/2020] [Accepted: 01/16/2020] [Indexed: 01/13/2023] Open
Abstract
Chromosome instability (CIN) has been associated with therapeutic resistance in many cancers. However, whether tumours become genomically unstable as an evolutionary mechanism to overcome the bottleneck exerted by therapy is not clear. Using a CIN model of Kras‐driven breast cancer, we demonstrate that aneuploid tumours acquire genetic modifications that facilitate the development of resistance to targeted therapy faster than euploid tumours. We further show that the few initially chromosomally stable cancers that manage to persist during treatment do so concomitantly with the acquisition of CIN. Whole‐genome sequencing analysis revealed that the most predominant genetic alteration in resistant tumours, originated from either euploid or aneuploid primary tumours, was an amplification on chromosome 6 containing the cMet oncogene. We further show that these tumours are dependent on cMet since its pharmacological inhibition leads to reduced growth and increased cell death. Our results highlight that irrespective of the initial CIN levels, cancer genomes are dynamic and the acquisition of a certain level of CIN, either induced or spontaneous, is a mechanism to circumvent oncogene addiction.
Collapse
Affiliation(s)
- Lorena Salgueiro
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Konstantina Rowald
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kalman Somogyi
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sridhar Kandala
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan O Korbel
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Rocio Sotillo
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
127
|
Bang Y, Su W, Schuler M, Nam D, Lim WT, Bauer TM, Azaro A, Poon RTP, Hong D, Lin C, Akimov M, Ghebremariam S, Zhao S, Giovannini M, Ma B. Phase 1 study of capmatinib in MET-positive solid tumor patients: Dose escalation and expansion of selected cohorts. Cancer Sci 2020; 111:536-547. [PMID: 31778267 PMCID: PMC7004521 DOI: 10.1111/cas.14254] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 01/01/2023] Open
Abstract
Capmatinib is an oral, ATP-competitive, and highly potent, type 1b MET inhibitor. Herein, we report phase 1 dose-escalation results for capmatinib in advanced MET-positive solid tumor patients and dose expansion in advanced non-lung tumors. Capmatinib was well tolerated with a manageable safety profile across all explored doses. Dose-limiting toxicities (DLT) occurred at 200 mg twice daily (bid), 250 mg bid, and 450 mg bid capsules; however, no DLT were reported at 600 mg bid (capsules). Capmatinib tablets at 400 mg bid had comparable tolerability and exposure to that of 600 mg bid capsules. Maximum tolerated dose was not reached; recommended phase 2 dose was 400 mg bid tablets/600 mg bid capsules; at this dose, Ctrough >EC90 (90% inhibition of c-MET phosphorylation in animal models) is expected to be achieved and maintained. Among the dose-expansion patients (N = 38), best overall response across all cohorts was stable disease (gastric cancer 22%, hepatocellular carcinoma 46%, other indications 28%); two other indication patients with gene copy number (GCN) ≥6 achieved substantial tumor reduction. Near-complete immunohistochemically determined phospho-MET inhibition (H-score = 2) was shown following capmatinib 450 mg bid capsule in paired biopsies obtained from one advanced colorectal cancer patient. Incidence of high-level MET GCN (GCN ≥6) and MET-overexpressing (immunohistochemistry 3+) tumors in the expansion cohorts was 8% and 13%, respectively; no MET mutations were observed. Thus, the recommended phase 2 dose (RP2D) of capmatinib was 600 mg bid capsule/400 mg bid tablet. Capmatinib was well tolerated and showed antitumor activity and acceptable safety profile at the RP2D. (ClinicalTrials.gov Identifier: NCT01324479).
Collapse
Affiliation(s)
- Yung‐Jue Bang
- Seoul National University College of MedicineSeoulKorea
| | - Wu‐Chou Su
- National Cheng Kung University HospitalTainanTaiwan
| | - Martin Schuler
- Department of Medical OncologyWest German Cancer CenterUniversity Duisburg‐Essen and German Cancer Consortium (DKTK), Partner Site University Hospital EssenEssenGermany
| | | | | | - Todd M. Bauer
- Sarah Cannon Research Institute/Tennessee OncologyPLLCNashvilleTennesseeUSA
| | - Analia Azaro
- Department of Medical OncologyMolecular Therapeutics Research UnitVall d’Hebron University HospitalBarcelonaSpain
- Pharmacology DepartmentThe Autonomous University of Barcelona(UAB)BarcelonaSpain
| | | | - David Hong
- University of Texas/MD Anderson Cancer CenterHoustonTexasUSA
| | | | | | | | - Sylvia Zhao
- China Novartis Institutes for BioMedical ResearchShanghaiChina
| | | | - Brigette Ma
- State Key Laboratory of Translational Oncology, Phase 1 Clinical Trial CentreThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
128
|
Carrigan G, Whipple S, Capra WB, Taylor MD, Brown JS, Lu M, Arnieri B, Copping R, Rothman KJ. Using Electronic Health Records to Derive Control Arms for Early Phase Single-Arm Lung Cancer Trials: Proof-of-Concept in Randomized Controlled Trials. Clin Pharmacol Ther 2020; 107:369-377. [PMID: 31350853 PMCID: PMC7006884 DOI: 10.1002/cpt.1586] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/21/2019] [Indexed: 11/11/2022]
Abstract
Oncology drug development increasingly relies on single-arm clinical trials. External controls (ECs) derived from electronic health record (EHR) databases may provide additional context. Patients from a US-based oncology EHR database were aligned with patients from randomized controlled trials (RCTs) and trial-specific eligibility criteria were applied to the EHR dataset. Overall survival (OS) in the EC-derived control arm was compared with OS in the RCT experimental arm. The primary outcome was OS, defined as time from randomization or treatment initiation (EHR) to death. Cox regression models were used to obtain effect estimates using EHR data. EC-derived hazard ratio estimates aligned closely with those from the corresponding RCT with one exception. Comparing log HRs among all RCT and EC results gave a Pearson correlation coefficient of 0.86. Properly selected control arms from contemporaneous EHR data could be used to put single-arm trials of OS in advanced non-small cell lung cancer into context.
Collapse
Affiliation(s)
| | | | | | | | - Jeffrey S. Brown
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Michael Lu
- GenentechSouth San FranciscoCaliforniaUSA
| | | | | | | |
Collapse
|
129
|
Kim D, Lee YS, Kim DH, Bae SC. Lung Cancer Staging and Associated Genetic and Epigenetic Events. Mol Cells 2020; 43:1-9. [PMID: 31999917 PMCID: PMC6999714 DOI: 10.14348/molcells.2020.2246] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022] Open
Abstract
The first step in treating lung cancer is to establish the stage of the disease, which in turn determines the treatment options and prognosis of the patient. Many factors are involved in lung cancer staging, but all involve anatomical information. However, new approaches, mainly those based on the molecular biology of cancer, have recently changed the paradigm for lung cancer treatment and have not yet been incorporated into staging. In a group of patients of the same stage who receive the same treatment, some may experience unexpected recurrence or metastasis, largely because current staging methods do not reflect the findings of molecular biological studies. In this review, we provide a brief summary of the latest research on lung cancer staging and the molecular events associated with carcinogenesis. We hope that this paper will serve as a bridge between clinicians and basic researchers and aid in our understanding of lung cancer.
Collapse
Affiliation(s)
- Dohun Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Chungbuk National University and Chungbuk National University Hospital, Cheongju 28644,
Korea
| | - You-Soub Lee
- Department of Biochemistry, College of Medicine, Chungbuk National University, Cheongju 28644,
Korea
| | - Duk-Hwan Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Suwon 16419,
Korea
| | - Suk-Chul Bae
- Department of Biochemistry, College of Medicine, Chungbuk National University, Cheongju 28644,
Korea
| |
Collapse
|
130
|
Chu QS. Targeting non-small cell lung cancer: driver mutation beyond epidermal growth factor mutation and anaplastic lymphoma kinase fusion. Ther Adv Med Oncol 2020; 12:1758835919895756. [PMID: 32047535 PMCID: PMC6984433 DOI: 10.1177/1758835919895756] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022] Open
Abstract
The identification of driver mutations in epidermal growth factor receptor, anaplastic lymphoma kinase, the BRAF and ROS1 genes and subsequent successful clinical development of kinase inhibitors not only significantly improves clinical outcomes but also facilitates the discovery of other novel driver mutations in non-small cell lung cancer. These driver mutations can be categorized into mutations in or near the kinase domain, gene amplification or fusion. In this review, BRAF V600E, EGFR and HER-2 exon 20 mutation, FGFR1-4, K-RAS, MET, neuregulin-1, NRTK, PI3K/AKT/mTOR, RET and ROS1 gene aberration and their therapeutics will be discussed.
Collapse
Affiliation(s)
- Quincy S. Chu
- Division of Medical Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta, T6G 1Z2, Canada
| |
Collapse
|
131
|
Genova C, Rossi G, Tagliamento M, Rijavec E, Biello F, Cerbone L, Zullo L, Grossi F. Targeted therapy of oncogenic-driven advanced non-small cell lung cancer: recent advances and new perspectives. Expert Rev Respir Med 2020; 14:367-383. [DOI: 10.1080/17476348.2020.1714441] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Carlo Genova
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giovanni Rossi
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Italy
| | - Marco Tagliamento
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Erika Rijavec
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Biello
- Oncology Unit, Ospedale Maggiore della Carità, Novara, Italy
| | - Luigi Cerbone
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lodovica Zullo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Grossi
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
132
|
A Randomized-Controlled Phase 2 Study of the MET Antibody Emibetuzumab in Combination with Erlotinib as First-Line Treatment for EGFR Mutation–Positive NSCLC Patients. J Thorac Oncol 2020; 15:80-90. [DOI: 10.1016/j.jtho.2019.10.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 11/21/2022]
|
133
|
Lung Carcinoma. Genomic Med 2020. [DOI: 10.1007/978-3-030-22922-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
134
|
Tafe LJ. Non-Small Cell Lung Cancer as a Precision Oncology Paradigm: Emerging Targets and Tumor Mutational Burden (TMB). Adv Anat Pathol 2020; 27:3-10. [PMID: 31567128 DOI: 10.1097/pap.0000000000000244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-small cell lung cancer (NSCLC), since the recognition of epidermal growth factor receptor (EGFR) mutations that sensitized tumors to EGFR tyrosine kinase inhibitors, has been a poster child for precision oncology in solid tumors. The emergence of resistance to the EGFR tyrosine kinase inhibitors led to the unveiling of multiple resistance mechanisms that are now recognized to be frequent mechanisms across multiple tumor types. Coevolution of technological advancements in testing methods available to clinical laboratories now has identified a growing number of molecularly defined subsets of NSCLC that have new therapeutic implications. In addition, identifying patients eligible for immunotherapy is another goal for precision oncology. Recently, studies suggest that TMB may be a promising biomarker for selecting patients with NSCLC for immunotherapy. This review focuses on emerging potentially targetable alterations specifically in RET, ERBB2 (HER2), MET, and KRAS and current evidence and controversies surrounding TMB testing.
Collapse
|
135
|
Moosavi F, Giovannetti E, Saso L, Firuzi O. HGF/MET pathway aberrations as diagnostic, prognostic, and predictive biomarkers in human cancers. Crit Rev Clin Lab Sci 2019; 56:533-566. [PMID: 31512514 DOI: 10.1080/10408363.2019.1653821] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/13/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022]
Abstract
Cancer is a major cause of death worldwide. MET tyrosine kinase receptor [MET, c-MET, hepatocyte growth factor (HGF) receptor] pathway activation is associated with the appearance of several hallmarks of cancer. The HGF/MET pathway has emerged as an important actionable target across many solid tumors; therefore, biomarker discovery becomes essential in order to guide clinical intervention and patient stratification with the aim of moving towards personalized medicine. The focus of this review is on how the aberrant activation of the HGF/MET pathway in tumor tissue or the circulation can provide diagnostic and prognostic biomarkers and predictive biomarkers of drug response. Many meta-analyses have shown that aberrant activation of the MET pathway in tumor tissue, including MET gene overexpression, gene amplification, exon 14 skipping and other activating mutations, is almost invariably associated with shorter survival and poor prognosis. Most meta-analyses have been performed in non-small cell lung cancer (NSCLC), breast, head and neck cancers as well as colorectal, gastric, pancreatic and other gastrointestinal cancers. Furthermore, several studies have shown the predictive value of MET biomarkers in the identification of patients who gain the most benefit from HGF/MET targeted therapies administered as single or combination therapies. The highest predictive values have been observed for response to foretinib and savolitinib in renal cancer, as well as tivantinib in NSCLC and colorectal cancer. However, some studies, especially those based on MET expression, have failed to show much value in these stratifications. This may be rooted in lack of standardization of methodologies, in particular in scoring systems applied in immunohistochemistry determinations or absence of oncogenic addiction of cancer cells to the MET pathway, despite detection of overexpression. Measurements of amplification and mutation aberrations are less likely to suffer from these pitfalls. Increased levels of MET soluble ectodomain (sMET) in circulation have also been associated with poor prognosis; however, the evidence is not as strong as it is with tissue-based biomarkers. As a diagnostic biomarker, sMET has shown its value in distinguishing cancer patients from healthy individuals in prostate and bladder cancers and in melanoma. On the other hand, increased circulating HGF has also been presented as a valuable prognostic and diagnostic biomarker in many cancers; however, there is controversy on the predictive value of HGF as a biomarker. Other biomarkers such as circulating tumor DNA (ctDNA) and tumor HGF levels have also been briefly covered. In conclusion, HGF/MET aberrations can provide valuable diagnostic, prognostic and predictive biomarkers and represent vital assets for personalized cancer therapy.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands
- Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza Onlus , Pisa , Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology, "Vittorio Erspamer," Sapienza University , Rome , Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
136
|
Kim Y, Bang SS, Jee S, Park S, Shin SJ, Jang K. Prevalence and Clinicopathological Significance of MET Overexpression and Gene Amplification in Patients with Gallbladder Carcinoma. Cancer Res Treat 2019; 52:481-491. [PMID: 31645095 PMCID: PMC7176974 DOI: 10.4143/crt.2019.370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/23/2019] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Mesenchymal epithelial transition (MET) is a proto-oncogene that encodes a heterodimeric transmembrane receptor tyrosine kinase for the hepatocyte growth factor. Aberrant MET signaling has been described in several solid tumors-especially non-small cell lung cancer- and is associated with tumor progression and adverse prognosis. As MET is a potential therapeutic target, information regarding its prevalence and clinicopathological relevance is crucial. MATERIALS AND METHODS We investigated MET expression and gene amplification in 113 gallbladder cancers using tissue microarray. Immunohistochemistry was used to evaluate MET overexpression, and silver/fluorescence in situ hybridization (ISH) was used to assess gene copy number. RESULTS MET overexpression was found in 37 cases of gallbladder carcinoma (39.8%), and gene amplification was present in 17 cases (18.3%). MET protein expression did not correlate with MET amplification. MET amplification was significantly associated with aggressive clinicopathological features, including high histological grade, advanced pT category, lymph node metastasis, and advanced American Joint Committee on Cancer stage. There was no significant correlation between any clinicopathological factors and MET overexpression. No difference in survival was found with respect to MET overexpression and amplification status. CONCLUSION Our data suggested that MET might be a potential therapeutic target for targeted therapy in gallbladder cancer, because MET amplification was found in a subset of tumors associated with adverse prognostic factors. Detection of MET amplification by ISH might be a useful predictive biomarker test for anti-MET therapy.
Collapse
Affiliation(s)
- Yeseul Kim
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Seong Sik Bang
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Seungyun Jee
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Sungeon Park
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Kiseok Jang
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
137
|
Lv PC, Yang YS, Wang ZC. Recent Progress in the Development of Small Molecule c-Met Inhibitors. Curr Top Med Chem 2019; 19:1276-1288. [PMID: 31526339 DOI: 10.2174/1568026619666190712205353] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/24/2019] [Accepted: 06/28/2019] [Indexed: 02/08/2023]
Abstract
C-Met, also referred to as Hepatocyte Growth Factor Receptor (HGFR), is a heterodimeric
receptor tyrosine kinase. It has been determined that c-Met gene mutations, overexpression, and amplification
also occur in a variety of human tumor types, and these events are closely related to the aberrant
activation of the HGF/c-Met signaling pathway. Meanwhile, high c-Met expression is closely associated
with poor prognosis in cancer patients. The c-Met kinase has emerged as an attractive target for developing
antitumor agents. In this review, we cover the recent advances on the small molecule c-Met inhibitors
discovered from 2018 until now, with a main focus on the rational design, synthesis and structureactivity
relationship analysis.
Collapse
Affiliation(s)
- Peng-Cheng Lv
- Department of Chemistry, Purdue University, West Lafayette, Indiana, IN 47907, United States
| | - Yu-Shun Yang
- Department of Chemistry, Purdue University, West Lafayette, Indiana, IN 47907, United States
| | - Zhong-Chang Wang
- Department of Chemistry, Purdue University, West Lafayette, Indiana, IN 47907, United States
| |
Collapse
|
138
|
Prell RA, Dybdal N, Arima A, Chihaya Y, Nijem I, Halpern W. Placental and Fetal Effects of Onartuzumab, a Met/HGF Signaling Antagonist, When Administered to Pregnant Cynomolgus Monkeys. Toxicol Sci 2019; 165:186-197. [PMID: 29893934 DOI: 10.1093/toxsci/kfy141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Onartuzumab is an engineered single arm, monovalent monoclonal antibody that targets the MET receptor and prevents hepatocyte growth factor (HGF) signaling. Knockout mice have clearly demonstrated that HGF/MET signaling is developmentally critical. A pre- and postnatal development study (enhanced design) was conducted in cynomolgus monkeys to evaluate the potential developmental consequences following onartuzumab administration. Control or onartuzumab, at loading/maintenance doses of 75/50 mg/kg (low) or 100/100 mg/kg (high), was administered intravenously once weekly to 12 confirmed pregnant female cynomolgus monkeys per group from gestation day (GD) 20 through GD 174. Onartuzumab administration resulted in decreased gestation length, decreased birth weight, and increased fetal and perinatal mortality. A GD147 C-section was conducted for a subset of Control and High Dose monkeys, and identified placental infarcts with hemorrhage in the chorionic plate, chorionic villus and/or decidual plate. These findings were limited to placentas from onartuzumab-treated animals. In addition, decreased cellularity of the hepatocytes with dilated hepatic sinusoids was inconsistently observed in the liver of a few fetal or infant monkeys that died in the perinatal period. Surviving offspring had some evidence of developmental delay compared with controls, but no overt teratogenicity. Overall, effects on the perinatal fetuses were consistent with those reported in knockout mice, but not as severe. Onartuzumab concentrations were low or below the level of detection in most offspring, with cord blood concentrations only 1%-2% of maternal levels on GD 147. Malperfusion secondary to onartuzumab-induced placental injury could explain the adverse pregnancy outcomes, fetal growth restriction and relatively low fetal exposures.
Collapse
Affiliation(s)
- Rodney A Prell
- Department of Safety Assessment, Genentech, South San Francisco, California 94080
| | - Noel Dybdal
- Department of Safety Assessment, Genentech, South San Francisco, California 94080
| | - Akihiro Arima
- Shin Nippon Biomedical Laboratories, Ltd, Kagoshima 891-1394, Japan
| | - Yutaka Chihaya
- Shin Nippon Biomedical Laboratories, Ltd., Atsubetsu-ku, Sapporo, Hokkaido 004-0071, Japan
| | - Ihsan Nijem
- Bioanalytical Sciences/Assay Development and Technology, Genentech, South San Francisco, California 94080
| | - Wendy Halpern
- Department of Safety Assessment, Genentech, South San Francisco, California 94080
| |
Collapse
|
139
|
Zhang C, Zhang J, Xu FP, Wang YG, Xie Z, Su J, Dong S, Nie Q, Shao Y, Zhou Q, Yang JJ, Yang XN, Zhang XC, Li Z, Wu YL, Zhong WZ. Genomic Landscape and Immune Microenvironment Features of Preinvasive and Early Invasive Lung Adenocarcinoma. J Thorac Oncol 2019; 14:1912-1923. [PMID: 31446140 DOI: 10.1016/j.jtho.2019.07.031] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/06/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Understanding the genomic landscape and immune microenvironment features of preinvasive and early invasive lung adenocarcinoma may provide critical insight and facilitate development of novel strategies for early detection and intervention. METHODS A total of 80 tumor tissue samples and 30 paired histologically normal lung tissue samples from 30 patients with adenocarcinoma in situ (AIS) (n = 8), minimally invasive adenocarcinoma (MIA) (n = 8), and invasive adenocarcinoma (IAC) (n = 14) were subjected to multiregion whole exome sequencing and immunohistochemistry staining for CD8 and programmed death ligand 1 (PD-L1). RESULTS All tumors, including AIS, exhibited evidence of genomic intratumor heterogeneity. Canonical cancer gene mutations in EGFR, erb-b2 receptor tyrosine kinase 2 gene (ERBB2), NRAS, and BRAF were exclusively trunk mutations detected in all regions within each tumor, whereas genes associated with cell mobility, gap junction, and metastasis were all subclonal mutations. EGFR mutation represented the most common driver alterations across AIS, MIA, and IAC, whereas tumor protein p53 gene (TP53) was identified in MIA and IAC but not in AIS. There was no difference in PD-L1 expression among AIS, MIA, and IAC, but the CD8 positivity rate was higher in IAC. Tumors positive for both PD-L1 and CD8 had a larger proportion of subclonal mutations. CONCLUSIONS Mutations in EGFR, ERBB2, NRAS, and BRAF are early clonal genomic events during carcinogenesis of lung adenocarcinoma, whereas TP53 and cell mobility, gap junction, and metastasis-related genes may be late events associated with subclonal diversification and neoplastic progression. Genomic intratumor heterogeneity and immunoediting are common and early phenomena that may have occurred before the acquisition of invasion.
Collapse
Affiliation(s)
- Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China; School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas; Department of Genomic Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Fang-Ping Xu
- Department of Pathology, Guangdong Provincial Peoples' Hospital, Guangzhou, People's Republic of China
| | - Yin-Guang Wang
- Nanjing Geneseeq Technology Inc., Najing, Jiangsu, People's Republic of China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Song Dong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Qiang Nie
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yang Shao
- Translational Medicine Research Institute, Geneseeq Technology Inc., Toronto, Ontario, Canada
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Zhi Li
- Department of Pathology, Guangdong Provincial Peoples' Hospital, Guangzhou, People's Republic of China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China.
| |
Collapse
|
140
|
Landi L, Chiari R, Tiseo M, D'Incà F, Dazzi C, Chella A, Delmonte A, Bonanno L, Giannarelli D, Cortinovis DL, de Marinis F, Borra G, Morabito A, Gridelli C, Galetta D, Barbieri F, Grossi F, Capelletto E, Minuti G, Mazzoni F, Verusio C, Bria E, Alì G, Bruno R, Proietti A, Fontanini G, Crinò L, Cappuzzo F. Crizotinib in MET-Deregulated or ROS1-Rearranged Pretreated Non–Small Cell Lung Cancer (METROS): A Phase II, Prospective, Multicenter, Two-Arms Trial. Clin Cancer Res 2019; 25:7312-7319. [DOI: 10.1158/1078-0432.ccr-19-0994] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/13/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
|
141
|
Del Re M, Crucitta S, Gianfilippo G, Passaro A, Petrini I, Restante G, Michelucci A, Fogli S, de Marinis F, Porta C, Chella A, Danesi R. Understanding the Mechanisms of Resistance in EGFR-Positive NSCLC: From Tissue to Liquid Biopsy to Guide Treatment Strategy. Int J Mol Sci 2019; 20:ijms20163951. [PMID: 31416192 PMCID: PMC6720634 DOI: 10.3390/ijms20163951] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
Liquid biopsy has emerged as an alternative source of nucleic acids for the management of Epidermal Growth Factor Receptor (EGFR)-mutant non-Small Cell Lung Cancer (NSCLC). The use of circulating cell-free DNA (cfDNA) has been recently introduced in clinical practice, resulting in the improvement of the identification of druggable EGFR mutations for the diagnosis and monitoring of response to targeted therapy. EGFR-dependent (T790M and C797S mutations) and independent (Mesenchymal Epithelial Transition [MET] gene amplification, Kirsten Rat Sarcoma [KRAS], Phosphatidyl-Inositol 4,5-bisphosphate 3-Kinase Catalytic subunit Alpha isoform [PI3KCA], and RAF murine sarcoma viral oncogene homolog B1 [BRAF] gene mutations) mechanisms of resistance to EGFR tyrosine kinase inhibitors (TKIs) have been evaluated in plasma samples from NSCLC patients using highly sensitive methods (i.e., digital droplet PCR, Next Generation Sequencing), allowing for the switch to other therapies. Therefore, liquid biopsy is a non-invasive method able to detect the molecular dynamic changes that occur under the pressure of treatment, and to capture tumor heterogeneity more efficiently than is allowed by tissue biopsy. This review addresses how liquid biopsy may be used to guide the choice of treatment strategy in EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giulia Gianfilippo
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology, 20141 Milano, Italy
| | - Iacopo Petrini
- General Pathology, Department of Translational Research & New Technologies in Surgery and Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giuliana Restante
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Angela Michelucci
- Unit of Molecular Genetics, Department of Laboratory Medicine, University Hospital, 56126 Pisa, Italy
| | - Stefano Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Filippo de Marinis
- Division of Thoracic Oncology, European Institute of Oncology, 20141 Milano, Italy
| | - Camillo Porta
- Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
- Division of Translational Oncology, I.R.C.C.S. Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy
| | - Antonio Chella
- Unit of Respiratory Medicine, Department of Critical Area and Surgical, Medical and Molecular Pathology, University Hospital, 56126 Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
142
|
Song X, Ji J, Gleason KJ, Yang F, Martignetti JA, Chen LS, Wang P. Insights into Impact of DNA Copy Number Alteration and Methylation on the Proteogenomic Landscape of Human Ovarian Cancer via a Multi-omics Integrative Analysis. Mol Cell Proteomics 2019; 18:S52-S65. [PMID: 31227599 PMCID: PMC6692782 DOI: 10.1074/mcp.ra118.001220] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 06/19/2019] [Indexed: 12/19/2022] Open
Abstract
In this work, we propose iProFun, an integrative analysis tool to screen for proteogenomic functional traits perturbed by DNA copy number alterations (CNAs) and DNA methylations. The goal is to characterize functional consequences of DNA copy number and methylation alterations in tumors and to facilitate screening for cancer drivers contributing to tumor initiation and progression. Specifically, we consider three functional molecular quantitative traits: mRNA expression levels, global protein abundances, and phosphoprotein abundances. We aim to identify those genes whose CNAs and/or DNA methylations have cis-associations with either some or all three types of molecular traits. Compared with analyzing each molecular trait separately, the joint modeling of multi-omics data enjoys several benefits: iProFun experienced enhanced power for detecting significant cis-associations shared across different omics data types, and it also achieved better accuracy in inferring cis-associations unique to certain type(s) of molecular trait(s). For example, unique associations of CNAs/methylations to global/phospho protein abundances may imply posttranslational regulations.We applied iProFun to ovarian high-grade serous carcinoma tumor data from The Cancer Genome Atlas and Clinical Proteomic Tumor Analysis Consortium and identified CNAs and methylations of 500 and 121 genes, respectively, affecting the cis-functional molecular quantitative traits of the corresponding genes. We observed substantial power gain via the joint analysis of iProFun. For example, iProFun identified 117 genes whose CNAs were associated with phosphoprotein abundances by leveraging mRNA expression levels and global protein abundances. By comparison, analyses based on phosphoprotein data alone identified none. A network analysis of these 117 genes revealed the known oncogene AKT1 as a key hub node interacting with many of the rest. In addition, iProFun identified one gene, BIN2, whose DNA methylation has cis-associations with its mRNA expression, global protein, and phosphoprotein abundances. These and other genes identified by iProFun could serve as potential drug targets for ovarian cancer.
Collapse
Affiliation(s)
- Xiaoyu Song
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jiayi Ji
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kevin J Gleason
- Department of Public Health Sciences, The University of Chicago, Chicago, IL
| | - Fan Yang
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - John A Martignetti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lin S Chen
- Department of Public Health Sciences, The University of Chicago, Chicago, IL.
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
143
|
Vansteenkiste JF, Van De Kerkhove C, Wauters E, Van Mol P. Capmatinib for the treatment of non-small cell lung cancer. Expert Rev Anticancer Ther 2019; 19:659-671. [PMID: 31368815 DOI: 10.1080/14737140.2019.1643239] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction: Activation of the MET pathway through MET amplifications or mutations is present in 3-4% of stage IV non-squamous non-small cell lung cancers (NSCLC). High MET amplifications and exon 14 skipping mutations are associated with poor prognosis: new treatments are needed for these patients. Capmatinib is a highly selective, potent small-molecule MET inhibitor with antitumor activity in NSCLC in vitro and in vivo. Areas covered: This article provides an overview of the capmatinib clinical development program in NSCLC, both as monotherapy in NSCLC with a dysregulated MET pathway, and in combination with epidermal growth factor receptor (EGFR) inhibitor therapy in EGFR-mutant NSCLC with MET-based acquired resistance to previous EGFR inhibition. Expert opinion: In the GEOMETRY Mono-1 study, treatment with capmatinib resulted in high response rates in stage IV NSCLC with MET exon 14 skipping mutations, particularly in first line, supporting testing for this biomarker at the time of diagnosis. Durable responses have been reported and results in MET-amplified NSCLC are eagerly anticipated. In EGFR-mutant NSCLC, notable responses have been observed in combination with an EGFR-tyrosine kinase inhibitor (TKI) in case of acquired resistance to EGFR-TKIs based on high MET amplification.
Collapse
Affiliation(s)
| | | | - Els Wauters
- Respiratory Oncology Unit (Respiratory Diseases), University Hospital KU Leuven , Leuven , Belgium
| | - Pierre Van Mol
- Respiratory Oncology Unit (Respiratory Diseases), University Hospital KU Leuven , Leuven , Belgium
| |
Collapse
|
144
|
Lung J, Hung MS, Lin YC, Lee KF, Jiang YY, Huang SL, Fang YH, Lu MS, Lin CK, Yang TM, Lin PY, Hsieh MJ, Tsai YH. MET exon 14 skipping mutations and gene amplification in a Taiwanese lung cancer population. PLoS One 2019; 14:e0220670. [PMID: 31369639 PMCID: PMC6675391 DOI: 10.1371/journal.pone.0220670] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/20/2019] [Indexed: 12/21/2022] Open
Abstract
Somatic mutations of MET gene are emerging as important driver mutations for lung cancers. To identify the common clinicopathological features of MET exon 14 skipping mutations and amplification and clarify whether the two MET gene alterations cause protein overexpression were investigated using 196 lung cancer samples of Taiwan through real time-qPCR/sequencing, fluorescence in situ hybridization, and immunohistochemistry. The two MET gene alterations are both present in low frequency, ~1%, in the studied lung cancer population of Taiwan. MET exon 14 skipping mutations were identified from two early-stage patients, who were both relatively advanced in age, and did not carry other driver mutations. One was an adenocarcinoma and the other was a rare carcinosarcoma. Three gene amplifications cases were identified. Neither of the two MET gene alterations would lead to protein overexpression; hence, direct detection in nucleic acid level would be a preferred and straightforward solution for the identification of skipping mutations. The presence of MET exon 14 mutations in minor histological types of lung cancers urge to extend screening scope of this mutation in lung cancer and treatment response evaluation in clinical trials. These would be important next steps for the success of MET target therapy in clinical practice.
Collapse
Affiliation(s)
- Jrhau Lung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Ming-Szu Hung
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Yu-Ching Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Kam-Fai Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Yuan Yuan Jiang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Shao-Lan Huang
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Yu-Hung Fang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Ming-Shian Lu
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Chin-Kuo Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Tsung-Ming Yang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Paul Yann Lin
- Department of Anatomic Pathology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Meng-Jer Hsieh
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Respiratory Care, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying Huang Tsai
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taiwan
- * E-mail:
| |
Collapse
|
145
|
Lu J, Li X, Tu K, Guan Y, Fung KP, Liu F. Verticillin A suppresses HGF-induced migration and invasion via repression of the c-Met/FAK/Src pathway in human gastric and cervical cancer cells. Onco Targets Ther 2019; 12:5823-5833. [PMID: 31440058 PMCID: PMC6668566 DOI: 10.2147/ott.s208683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/12/2019] [Indexed: 01/06/2023] Open
Abstract
Background and purpose: Verticillin A is a fungal epipolythiodioxopiperazine (ETP) metabolite that was isolated from Amanita flavorubescens Alk infected by Verticillium sp. It was previously proven to possess potent anti-tumor cell growth activity, and we have recently determined that verticillin A is a selective inhibitor of H3K9me3-specific histone methyltransferase. The objective of this study was to find out whether verticillin A is an effective agent for suppression of gastric and cervical tumor progression. Materials and methods: Wound healing and transwell assays was performed to evaluate the effect of verticillin A on hepatocyte growth factor (HGF)-induced AGS and HeLa cells migration and invasion in vitro. Western blot was used to detect signaling proteins verticillin A affected. Results: We determined that verticillin A effectively suppressed hepatocyte growth factor (HGF)-induced AGS and HeLa cells migration and invasion in vitro. At the molecular level, we demonstrated that verticillin A inhibited HGF-induced c-Met phosphorylation and repressed the expression of total c-Met protein in AGS and HeLa cells, resulting from reduced expression of fatty acid synthase. In addition, verticillin A could suppress c-Met downstream FAK/Src signaling pathways by impairing c-Met phosphorylation induced by HGF. Conclusion: Our study demonstrated verticillin A inhibits the migration ability of human gastric cancer (AGS) cells and cervical cancer (HeLa) cells by targeting c-Met and its downstream FAK/Src signaling pathways, and suggested that verticillin A acts as a novel HGF/c-Met inhibitor by reducing expression of this receptor.
Collapse
Affiliation(s)
- Jingxin Lu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xia Li
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou 310022, People’s Republic of China
| | - Kai Tu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yuelin Guan
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| | - Kwok-Pui Fung
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
- School of Biomedical Sciences (SBS), The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Feiyan Liu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
146
|
Design, synthesis and evaluation of sulfonylurea-containing 4-phenoxyquinolines as highly selective c-Met kinase inhibitors. Bioorg Med Chem 2019; 27:2801-2812. [DOI: 10.1016/j.bmc.2019.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/26/2019] [Accepted: 05/04/2019] [Indexed: 12/20/2022]
|
147
|
Synthesis and anti-tumor activity of [1,4] dioxino [2,3-f] quinazoline derivatives as dual inhibitors of c-Met and VEGFR-2. Bioorg Chem 2019; 88:102916. [DOI: 10.1016/j.bioorg.2019.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 11/20/2022]
|
148
|
Tsakonas G, Botling J, Micke P, Rivard C, LaFleur L, Mattsson J, Boyle T, Hirsch FR, Ekman S. c-MET as a biomarker in patients with surgically resected non-small cell lung cancer. Lung Cancer 2019; 133:69-74. [DOI: 10.1016/j.lungcan.2019.04.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/14/2019] [Accepted: 04/30/2019] [Indexed: 01/22/2023]
|
149
|
Riedel R, Michels S, Heydt C, Siemanowski J, Kobe C, Bunck A, Schäfer S, Fischer RN, Scheffler M, Abdulla DS, Nogová L, Koleczko S, Merkelbach-Bruse S, Büttner R, Wolf J. Acquired KRAS mutation and loss of low-level MET amplification after durable response to crizotinib in a patient with lung adenocarcinoma. Lung Cancer 2019; 133:20-22. [DOI: 10.1016/j.lungcan.2019.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/01/2019] [Accepted: 05/05/2019] [Indexed: 11/28/2022]
|
150
|
Wang F, Lu JB, Wu XY, Feng YF, Shao Q, An X, Wang HY. Clinical genetic features and related survival implications in patients with surgically resected large-cell lung cancer. Cancer Manag Res 2019; 11:5489-5499. [PMID: 31354355 PMCID: PMC6585161 DOI: 10.2147/cmar.s200263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/10/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Large-cell lung carcinomas (LCLCs) were reclassified by the World Health Organization 2015 criteria. and remain fairly unknown at the molecular level and targeted-therapeutic options. Methods: Data of 184 lung cancer patients were retrieved from clinical records, of which 54 were found to be pathologically diagnosed as LCLC. The genetic alterations EGFR/KRAS/BRAF mutations, MET copy number, and exon 14 mutation, ALK and ROS1 rearrangements, and PDL1 expression were investigated using clinical technologies. The relationship between clinicopathologic and genetic features was analyzed, and the Kaplan–Meier method with log-rank test was used for analyzing patient survival. Results: Major events, including EGFR, KRAS, and BRAF mutations and MET copy-number gain, were found in 5.6%, 16.7%, 1.9%, and 18.5% in LCLC, respectively. No ALK or ROS1 translocation was detected. PDL1 expression in tumor cells and in tumor-infiltrating lymphocytes was observed in 24 (44.4%) and 16 (29.6%) patients. Kaplan–Meier analysis showed that patients with a KRAS mutation had ower 5-year overall survival than those with wild-type KRAS (25.4% vs 47.8%, P=0.028) and that patients with negative PDL1 stained in tumor cells but positive for tumor-infiltrating lymphocytes had significantly favorable overall survival compared to those with solitary and positive PDL1 stained in tumor cells (62.5% vs 20.6%, P=0.044). Conclusion:KRAS mutations and PDL1 expression can predict patient survival and be potential target options in LCLC.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jia-Bin Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Xiao-Yan Wu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Yan-Fen Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qiong Shao
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Xin An
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Hai-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| |
Collapse
|