201
|
Sun CC, Li SJ, Zhang F, Pan JY, Wang L, Yang CL, Xi YY, Li DJ. Hsa-miR-329 exerts tumor suppressor function through down-regulation of MET in non-small cell lung cancer. Oncotarget 2017; 7:21510-26. [PMID: 26909600 PMCID: PMC5008302 DOI: 10.18632/oncotarget.7517] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/05/2016] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) act as key regulators of multiple cancers. Hsa-miR-329 (miR-329) functions as a tumor suppressor in some malignancies. However, its role on lung cancer remains poorly understood. In this study, we investigated the role of miR-329 on the development of lung cancer. The results indicated that miR-329 was decreased in primary lung cancer tissues compared with matched adjacent normal lung tissues and very low levels were found in a non-small cell lung cancer (NSCLC) cell lines. Ectopic expression of miR-329 in lung cancer cell lines substantially repressed cell growth as evidenced by cell viability assay, colony formation assay and BrdU staining, through inhibiting cyclin D1, cyclin D2 and up-regulatiing p57(Kip2) and p21(WAF1/CIP1). In addition, miR-329 promoted NSCLC cell apoptosis, as indicated by up-regulation of key apoptosis gene cleaved caspase-3, and down-regulation of anti-apoptosis gene Bcl2. Moreover, miR-329 inhibited cellular migration and invasiveness through inhibiting matrix metalloproteinases (MMP)-7 and MMP-9. Further, oncogene MET was revealed to be a putative target of miR-329, which was inversely correlated with miR-329 expression. Furthermore, down-regulation of MET by siRNA performed similar effects to over-expression of miR-329. Collectively, our results demonstrated that miR-329 played a pivotal role in lung cancer through inhibiting cell proliferation, migration, invasion, and promoting apoptosis by targeting oncogenic MET.
Collapse
Affiliation(s)
- Cheng-Cao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| | - Shu-Jun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China.,Wuhan Hospital for The Prevention and Treatment of Occupational Diseases, Wuhan, P. R. China
| | - Feng Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| | - Jing-Yu Pan
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| | - Liang Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| | - Cui-Li Yang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| | - Yong-Yong Xi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| | - De Jia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
202
|
Angevin E, Spitaleri G, Rodon J, Dotti K, Isambert N, Salvagni S, Moreno V, Assadourian S, Gomez C, Harnois M, Hollebecque A, Azaro A, Hervieu A, Rihawi K, De Marinis F. A first-in-human phase I study of SAR125844, a selective MET tyrosine kinase inhibitor, in patients with advanced solid tumours with MET amplification. Eur J Cancer 2017; 87:131-139. [PMID: 29145039 DOI: 10.1016/j.ejca.2017.10.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE Dysregulated MET signalling is implicated in oncogenesis. The safety and preliminary efficacy of a highly selective MET kinase inhibitor (SAR125844) was investigated in patients with advanced solid tumours and MET dysregulation. METHODS This was a phase I dose-escalation (3 + 3 design [50-740 mg/m2]) and dose-expansion study. In the dose escalation, patients had high total MET (t-MET) expression by immunohistochemistry (IHC) or MET amplification by fluorescence in situ hybridisation. In the dose expansion, patients had MET amplification (including a subset of patients with non-small cell lung cancer [NSCLC]) or phosphorylated-MET (p-MET) expression (IHC). Objectives were determination of maximum tolerated dose (MTD) of once-weekly intravenous SAR125844 based on dose-limiting toxicities; safety and pharmacokinetic profile; preliminary efficacy of SAR125844 MTD in the expansion cohort. RESULTS In total, 72 patients were enrolled: dose escalation, N = 33; dose expansion, N = 39; 570 mg/m2 was established as the MTD. Most frequent treatment-emergent adverse events (AEs) were asthenia/fatigue (58.3%), nausea (31.9%), and abdominal pain, constipation, and dyspnea (27.8% for each); 58.3% of patients reported grade 3 AEs (19.4% were treatment related). Of the 29 evaluable patients with MET amplification treated at 570 mg/m2, five achieved a partial response, including four of 22 with NSCLC; 17 patients had stable disease. No response was observed in patients with high p-MET solid tumours. There was no correlation between tumour response and t-MET status or MET gene copy number. CONCLUSION The MTD of once-weekly SAR125844 was 570 mg/m2; SAR125844 was well tolerated, with significant antitumour activity in patients with MET-amplified NSCLC. CLINICAL TRIAL REGISTRATION NUMBER NCT01391533.
Collapse
Affiliation(s)
- Eric Angevin
- Drug Development Department, Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Université Paris-Saclay, Gustave Roussy, Villejuif, F-94805, France.
| | - Gianluca Spitaleri
- Thoracic Oncology Division, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy.
| | - Jordi Rodon
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, P. Vall d'Hebron 119-129, Barcelona, 08035, Spain.
| | - Katia Dotti
- Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy.
| | - Nicolas Isambert
- Centre Georges-François Leclerc, 1 Rue du Professeur Marion, 21000, Dijon, France.
| | - Stefania Salvagni
- Oncologia Medica, S. Orsola-Malpighi University Hospital Bologna, Via Pietro Albertoni, 15, 40138, Bologna, Italy.
| | - Victor Moreno
- START MADRID - FJD., Hospital Universitario Fundación Jiménez Díaz, vda. Reyes Católicos, 2, 28040, Madrid, Spain.
| | | | | | | | - Antoine Hollebecque
- Drug Development Department, Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Université Paris-Saclay, Gustave Roussy, Villejuif, F-94805, France.
| | - Analia Azaro
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, P. Vall d'Hebron 119-129, Barcelona, 08035, Spain.
| | - Alice Hervieu
- Centre Georges-François Leclerc, 1 Rue du Professeur Marion, 21000, Dijon, France.
| | - Karim Rihawi
- Oncologia Medica, S. Orsola-Malpighi University Hospital Bologna, Via Pietro Albertoni, 15, 40138, Bologna, Italy.
| | - Filippo De Marinis
- Thoracic Oncology Division, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy.
| |
Collapse
|
203
|
Genetic screening and molecular characterization of MET alterations in non-small cell lung cancer. Clin Transl Oncol 2017; 20:881-888. [DOI: 10.1007/s12094-017-1799-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/05/2017] [Indexed: 11/26/2022]
|
204
|
Zhang J, Sun J, Zhang Z, Liang X, Luo Y, Wu S, Liang Z. Protein overexpression and gene amplification of cellular mesenchymal-epithelial transition factor is associated with poor prognosis in micropapillary-predominant subtype pulmonary adenocarcinoma. Hum Pathol 2017; 72:59-65. [PMID: 29128478 DOI: 10.1016/j.humpath.2017.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/22/2017] [Accepted: 10/27/2017] [Indexed: 11/26/2022]
Abstract
Micropapillary-predominant subtype pulmonary adenocarcinoma (MPPAC) is a subtype of lung cancer with poor prognosis. Cellular mesenchymal-epithelial transition factor (c-MET) is a promising pharmaceutic target found to be associated with the survival of patients with pulmonary adenocarcinoma. In this study, we aimed to analyze c-MET protein overexpression and gene amplification in MPPAC samples and to elucidate their relationship with the clinicopathological characteristics of the patients. c-MET protein expression was examined by immunohistochemical analyses, and gene amplification was detected by fluorescence in situ hybridization. A total of 86 MPPAC cases were included in this study. The prevalence of c-MET protein overexpression and gene amplification were 62.8% and 10.5%, respectively. C-MET protein overexpression was significantly associated with smoking status, lymphatic and venous invasion, and tumor-node-metastasis stage (P = .014, P = .040, and P = .004, respectively), but c-MET gene amplification showed no relation with any of these characteristics. Univariate analysis revealed that pleural invasion, lymph node metastasis, lymphatic and venous invasion, tumor-node-metastasis stage, c-MET protein overexpression, and c-MET gene amplification were associated with poor prognosis (P = .041, P < .001, P = .001, P < .001, P = .001 and P = .001, respectively), but only c-MET gene amplification was an independent prognostic marker (P = .04). These results indicated that c-MET is an important biomarker. Also, c-MET protein overexpression and gene amplification are highly related to poor prognosis in patients with MPPAC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730
| | - Jian Sun
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730
| | - Zhiwen Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730
| | - Xiaolong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730
| | - Yufeng Luo
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730
| | - Shafei Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China, 100730.
| |
Collapse
|
205
|
Abstract
The expanding spectrum of both established and candidate oncogenic driver mutations identified in non-small-cell lung cancer (NSCLC), coupled with the increasing number of clinically available signal transduction pathway inhibitors targeting these driver mutations, offers a tremendous opportunity to enhance patient outcomes. Despite these molecular advances, advanced-stage NSCLC remains largely incurable due to therapeutic resistance. In this Review, we discuss alterations in the targeted oncogene ('on-target' resistance) and in other downstream and parallel pathways ('off-target' resistance) leading to resistance to targeted therapies in NSCLC, and we provide an overview of the current understanding of the bidirectional interactions with the tumour microenvironment that promote therapeutic resistance. We highlight common mechanistic themes underpinning resistance to targeted therapies that are shared by NSCLC subtypes, including those with oncogenic alterations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), ROS1 proto-oncogene receptor tyrosine kinase (ROS1), serine/threonine-protein kinase b-raf (BRAF) and other less established oncoproteins. Finally, we discuss how understanding these themes can inform therapeutic strategies, including combination therapy approaches, and overcome the challenge of tumour heterogeneity.
Collapse
Affiliation(s)
- Julia Rotow
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, 505 Parnassus Avenue, Box 1270, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0981, San Francisco, California 94143, USA
| | - Trever G Bivona
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, 505 Parnassus Avenue, Box 1270, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0981, San Francisco, California 94143, USA
- Cellular and Molecular Pharmacology, University of California San Francisco, Box 2140, San Francisco, California 94158, USA
| |
Collapse
|
206
|
Baldacci S, Mazieres J, Tomasini P, Girard N, Guisier F, Audigier-Valette C, Monnet I, Wislez M, Pérol M, Dô P, Dansin E, Leduc C, Giroux Leprieur E, Moro-Sibilot D, Tulasne D, Kherrouche Z, Labreuche J, Cortot AB. Outcome of EGFR-mutated NSCLC patients with MET-driven resistance to EGFR tyrosine kinase inhibitors. Oncotarget 2017; 8:105103-105114. [PMID: 29285237 PMCID: PMC5739624 DOI: 10.18632/oncotarget.21707] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Background Several mechanisms of acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) in EGFR-mutated NSCLC have been described including the T790M mutation and MET amplification. Whereas T790M mutation confers prolonged survival and sensitivity to 3rd generation TKIs, data are lacking on clinical features and outcome of MET-driven resistant EGFR-mutated NSCLC patients. Methods Patients with metastatic EGFR-mutated NSCLC displaying high MET overexpression or MET amplification, detected on a biopsy performed after progression on EGFR TKI, were identified in 15 centers. Clinical and molecular data were retrospectively collected. Results Forty two patients were included. The median overall survival (OS), and the median post EGFR TKI progression overall survival (PPOS) were 36.2 months [95%CI 27.3-66.5] and 18.5 months [95%CI 10.6-27.4] respectively. Nineteen out of 36 tumors tested for MET FISH had MET amplification. A T790M mutation was found in 11/41 (26.8%) patients. T790M-positive patients had a better OS than T790M-negative patients (p=0.0224). Nineteen patients received a MET TKI. Objective response was reported in 1 out of 12 evaluable patients treated with a MET inhibitor as a single agent and in 1 of 2 patients treated with a combination of MET and EGFR TKIs. Conclusion MET-driven resistance to EGFR TKI defines a specific pattern of resistance characterized by low objective response rate to MET inhibitors given alone and overlapping with T790M mutations. Further studies are warranted to define adequate therapeutic strategies for MET-driven resistance to EGFR TKI.
Collapse
Affiliation(s)
- Simon Baldacci
- CHU Lille, Thoracic Oncology Department, Univ. Lille, Siric ONCOLille, Lille, France.,Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161, M3T, Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | - Julien Mazieres
- Toulouse University Hospital, Université Paul Sabatier, Toulouse, France
| | - Pascale Tomasini
- Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Multidisciplinary Oncology & Therapeutic Innovations Department, Marseille, France
| | - Nicolas Girard
- Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | - Florian Guisier
- Rouen University Hospital, Thoracic oncology unit & Normandy University, IRIB, LITIS Lab, EA 4103 QuantIF team, Rouen, France
| | | | - Isabelle Monnet
- Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | | | - Maurice Pérol
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Pascal Dô
- Centre Régional de Lutte Contre le Cancer François Baclesse, Caen, France
| | | | | | | | - Denis Moro-Sibilot
- Unité d'Oncologie Thoracique, Service de Pneumologie, CHU Grenoble-Alpes, La Tronche, France
| | - David Tulasne
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161, M3T, Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | - Zoulika Kherrouche
- CHU Lille, Thoracic Oncology Department, Univ. Lille, Siric ONCOLille, Lille, France.,Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161, M3T, Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | | | - Alexis B Cortot
- CHU Lille, Thoracic Oncology Department, Univ. Lille, Siric ONCOLille, Lille, France.,Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161, M3T, Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| |
Collapse
|
207
|
Kim JH, Kim HS, Kim BJ. MET inhibitors in advanced non-small-cell lung cancer: a meta-analysis and review. Oncotarget 2017; 8:75500-75508. [PMID: 29088885 PMCID: PMC5650440 DOI: 10.18632/oncotarget.20824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/27/2017] [Indexed: 12/19/2022] Open
Abstract
The alterations of MET have been detected in non-small-cell lung cancer (NSCLC). However, survival benefit of MET inhibitors remains controversial. We performed this meta-analysis to evaluate the survival benefit of MET inhibitors combined with an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) or standard chemotherapy in patients with advanced or metastatic NSCLC. A systematic computerized search of the electronic databases was carried out. From seven studies, 2,577 patients were included in the meta-analysis. Compared with patients in the placebo group, patients who received an additional MET inhibitor did not show significantly improved progression-free survival (hazard ration (HR) = 0.92 [95% confidence interval (CI): 0.79–1.08], P = 0.33) and overall survival (HR = 1.0 [95% CI: 0.90–1.11], P = 0.97). In the subgroup analysis, patients with MET-high NSCLC tended to show longer survival when treated with an additional MET inhibitor than those in the placebo group (HR = 0.76, [95% CI: 0.58–1.01], P = 0.06). In conclusion, this meta-analysis indicates that the addition of a MET inhibitor to an EGFR TKI or chemotherapy has no survival benefit over placebo in patients with advanced or metastatic NSCLC. Although patients with MET-high tumor tended to show better survival, further studies to explore more specific biomarkers are warranted to identify ideal candidates for MET inhibitors in NSCLC.
Collapse
Affiliation(s)
- Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Bum Jun Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea.,Department of Internal Medicine, National Army Capital Hospital, The Armed Forces Medical Command, Sungnam 13574, Republic of Korea
| |
Collapse
|
208
|
Lau KW, Seng C, Lim TKH, Tan DSW. Expanded molecular interrogation for potential actionable targets in non-squamous non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:372. [PMID: 29057232 PMCID: PMC5635263 DOI: 10.21037/atm.2017.08.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/03/2017] [Indexed: 12/15/2022]
Abstract
The advent of targeted therapies has established new standards of care for defined molecular subsets of non-small cell lung cancer (NSCLC). Not only has this led to significant changes in the routine clinical management of lung cancer e.g., multiplexed genomic testing, but it has provided important principles and benchmarks for determining "actionability". At present, the clinical paradigms are most evolved for EGFR mutations and ALK rearrangements, where multiple randomized phase III trials have determined optimal treatment strategies in both treatment naïve and resistant settings. However, this may not always be feasible with low prevalence alterations e.g., ROS1 and BRAF mutations. Another emerging observation is that not all targets are equally "actionable", necessitating a rigorous preclinical, clinical and translational framework to prosecute new targets and drug candidates. In this review, we will cover the role of targeted therapies for NSCLC harbouring BRAF, MET, HER2 and RET alterations, all of which have shown promise in non-squamous non-small cell lung cancer (ns-NSCLC). We further review some early epigenetic targets in NSCLC, an area of emerging interest. With increased molecular segmentation of lung cancer, we discuss the upcoming challenges in drug development and implementation of precision oncology approaches, especially in light of the complex and rapidly evolving therapeutic landscape.
Collapse
Affiliation(s)
- Kah Weng Lau
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
- Institute of Molecular and Cell Biology, ASTAR, Singapore
| | - Claudia Seng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Tony K H Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer Therapeutics Research Laboratory, Singapore
- Genome Institute of Singapore, ASTAR, Singapore
| |
Collapse
|
209
|
Xu C, Wang W, Wu M, Zhu Y, Zhuang W, Lin G, Du K, Huang Y, Chen Y, Chen G, Fang M. Comparison of the c-MET gene amplification between primary tumor and metastatic lymph nodes in non-small cell lung cancer. Thorac Cancer 2017; 8:417-422. [PMID: 28590585 PMCID: PMC5582467 DOI: 10.1111/1759-7714.12455] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND c-MET has recently been identified as a promising novel target in non-small cell lung cancer (NSCLC). We detected the consistency of c-MET gene amplification in metastatic lymph nodes and tumor tissues of NSCLC patients and discuss the clinical application value of c-MET gene amplification in metastatic lymph nodes. METHODS Real-time fluorescent quantitative PCR was used to test tumor tissues in 368 NSCLC patients and 178 paired metastatic lymph node samples. The amplification consistency in metastatic lymph nodes and tissue samples were compared and the correlation between c-MET gene amplification and the clinical characteristics of patients was analyzed. RESULTS The c-MET gene amplification rate was 8.97% (33/368) in tumor tissues. Of the 178 paired cases, c-MET gene amplification was positive in 7.95% (15/178) of cancerous tissues and 18.54% (33/178) of metastatic lymph nodes. c-MET gene amplification was detected more frequently in metastatic lymph nodes than in primary cancerous tissue. When metastatic lymph nodes were used as surrogate samples of primary cancerous tissues, the sensitivity was 86.67% (13/15) and the specificity was 87.69% (143/163). CONCLUSIONS Screening for c-MET gene amplification in lymph node metastases could determine which patients are eligible for tyrosine kinase inhibitor therapy. Lymph node metastasis can predict c-MET gene amplification in a primary tumor and guide the clinical use of c-MET gene targeted drugs.
Collapse
Affiliation(s)
- Chun‐wei Xu
- Department of Pathology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Wen‐xian Wang
- Department of ChemotherapyZhejiang Cancer HospitalHangzhouChina
| | - Mei‐juan Wu
- Department of PathologyZhejiang Cancer HospitalHangzhouChina
| | - You‐cai Zhu
- Department of Thoracic Disease CenterZhejiang Rongjun HospitalJiaxingChina
| | - Wu Zhuang
- Department of Medical Thoracic Oncology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Gen Lin
- Department of Medical Thoracic Oncology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Kai‐qi Du
- Department of Thoracic Disease CenterZhejiang Rongjun HospitalJiaxingChina
| | - Yun‐jian Huang
- Department of Medical Thoracic Oncology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Yan‐ping Chen
- Department of Pathology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Gang Chen
- Department of Pathology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Mei‐yu Fang
- Department of Comprehensive Medical OncologyZhejiang Cancer HospitalHangzhouChina
| |
Collapse
|
210
|
Pilotto S, Carbognin L, Karachaliou N, Ma PC, Rosell R, Tortora G, Bria E. Tracking MET de-addiction in lung cancer: A road towards the oncogenic target. Cancer Treat Rev 2017; 60:1-11. [PMID: 28843992 DOI: 10.1016/j.ctrv.2017.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/05/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023]
Abstract
The discovery of druggable oncogenic drivers (i.e. EGFR and ALK), along with the introduction of comprehensive tumor genotyping techniques into the daily clinical practice define non-small-cell lung cancer (NSCLC) asa group of heterogeneous diseases, requiring a context-personalized clinico-therapeutical approach. Among the most investigated biomarkers, the MET proto-oncogene has been extensively demonstrated to play a crucial role throughout the lung oncogenesis, unbalancing the proliferation/apoptosis signaling and influencing the epithelial-mesenchymal transition and the invasive phenotype. Nevertheless, although different mechanisms eliciting the aberrant MET-associated oncogenic stimulus have been detected in lung cancer (such as gene amplification, increased gene copy number, mutations and MET/HGF overexpression), to date no clinically impactful results have been achieved with anti-MET tyrosine kinase inhibitors and monoclonal antibodies in the context of an unselected or MET enriched population. Recently, MET exon 14 splicing abnormalities have been identified asa potential oncogenic target in lung cancer, able to drive the activity of MET inhibitors in molecularly selected patients. In this paper, the major advancement and drawbacks of MET history in lung cancer are reviewed, underlying the renewed scientific euphoria related to the recent identification of MET exon 14 splicing variants asan actionable oncogenic target.
Collapse
Affiliation(s)
- S Pilotto
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | - L Carbognin
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | | | - P C Ma
- WVU Cancer Institute, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, United States; WV Clinical and Translational Science Institute, Morgantown, WV, United States.
| | - R Rosell
- Pangaea Biotech, Barcelona, Spain; Instituto Oncológico Dr Rosell, Quiron-Dexeus University Hospital, Barcelona, Spain; Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Spain; Molecular Oncology Research (MORe) Foundation, Barcelona, Spain; Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Spain.
| | - G Tortora
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | - E Bria
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| |
Collapse
|
211
|
Chan KI, Vong HT, Sin LF, Yip YC, Zhong XY, Wen JM. Relationship between driver gene mutations, their relative protein expressions and survival in non-small cell lung carcinoma in Macao. CLINICAL RESPIRATORY JOURNAL 2017; 12:1416-1423. [PMID: 28756651 DOI: 10.1111/crj.12670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/23/2017] [Accepted: 07/25/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVES We report the status of most common gene mutations in non-small cell lung carcinoma (NSCLC) in Macao, and explore the relationship between each gene mutation and clinicopathologic features and survival. METHODS EGFR, KRAS and BRAF mutations were detected by PCR in 122 cases of NSCLC. ALK translocation and MET amplification were detected by fluorescence in situ hybridization (FISH). MET and thyroid transcription factor (TTF-1) were investigated by immunohistochemistry. Clinical data were collected for analyzing their correlation with the gene mutations. RESULTS The mutation of EGFR, KRAS and BRAF was detected in 48 (39.3%), 13 (10.7%) and 3 (2.5%) of 122 cases of NSCLC, respectively. ALK translocation and MET amplification were detected in 7 (5.7%) and 3 cases (2.5%). The rate of EGFR mutation was significantly higher in female and non-smoker patients. In TTF-1 positive cases EGFR mutation was more frequent. Age of the patients over 62-year old was correlated with KRAS mutations. The concordance between ALK IHC and FISH was 58.3%. The MET protein in the cases with MET amplification was 100% positive. The survival was lower in the patients with positive MET protein than those with negative. MET protein was an independent prognostic factor for NSCLC. CONCLUSIONS EGFR mutation occurred frequently in the female never smoke patients with NSCLC. KRAS mutation was more common in old patients. Negative MET protein expression could be used as a negative predictive marker of MET amplification. MET protein expression was an independent prognostic factor for NSCLC.
Collapse
Affiliation(s)
- Kin Iong Chan
- Department of Pathology, Kiang Wu Hospital, Macau Special Administrative Region, Macau, China
| | - Hong Ting Vong
- Department of Pathology, Kiang Wu Hospital, Macau Special Administrative Region, Macau, China
| | - Lai Fong Sin
- Department of Pathology, Kiang Wu Hospital, Macau Special Administrative Region, Macau, China
| | - Yuk Ching Yip
- Department of Pathology, Kiang Wu Hospital, Macau Special Administrative Region, Macau, China
| | - Xue Yun Zhong
- Department of Pathology, Medical School, Jinan University, Guangzhou 510632, China
| | - Jian Ming Wen
- Department of Pathology, Kiang Wu Hospital, Macau Special Administrative Region, Macau, China
| |
Collapse
|
212
|
Salgia R. MET in Lung Cancer: Biomarker Selection Based on Scientific Rationale. Mol Cancer Ther 2017; 16:555-565. [PMID: 28373408 DOI: 10.1158/1535-7163.mct-16-0472] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/21/2016] [Accepted: 12/19/2016] [Indexed: 11/16/2022]
Abstract
MET or hepatocyte growth factor (HGF) receptor pathway signaling mediates wound healing and hepatic regeneration, with pivotal roles in embryonic, neuronal, and muscle development. However, dysregulation of MET signaling mediates proliferation, apoptosis, and migration and is implicated in a number of malignancies. In non-small cell lung cancer (NSCLC), aberrant MET signaling can occur through a number of mechanisms that collectively represent a significant proportion of patients. These include MET or HGF protein overexpression, MET gene amplification, MET gene mutation or fusion/rearrangement, or aberrations in downstream signaling or regulatory components. Responses to MET tyrosine kinase inhibitors have been documented in clinical trials in patients with MET-amplified or MET-overexpressing NSCLC, and case studies or case series have shown that MET mutation/deletion is a biomarker that is also predictive of response to these agents. However, other recent clinical data have highlighted an urgent need to elucidate optimal biomarkers based on genetic and/or protein diagnostics to correctly identify patients most likely to benefit in ongoing clinical trials of an array of MET-targeted therapies of differing class. The latest advances in the development of MET biomarkers in NSCLC have been reviewed, toward establishing appropriate MET biomarker selection based on a scientific rationale. Mol Cancer Ther; 16(4); 555-65. ©2017 AACR.
Collapse
Affiliation(s)
- Ravi Salgia
- City of Hope, Department of Medical Oncology and Therapeutics Research, Duarte, California.
| |
Collapse
|
213
|
Al-Saad S, Richardsen E, Kilvaer TK, Donnem T, Andersen S, Khanehkenari M, Bremnes RM, Busund LT. The impact of MET, IGF-1, IGF1R expression and EGFR mutations on survival of patients with non-small-cell lung cancer. PLoS One 2017; 12:e0181527. [PMID: 28742836 PMCID: PMC5526580 DOI: 10.1371/journal.pone.0181527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/03/2017] [Indexed: 01/10/2023] Open
Abstract
Introduction To compare the efficacy of silver in situ hybridization (SISH) and immunohistochemistry (IHC) in detecting MET and IGF1R alterations and to investigate their prevalence and prognostic significance. A possible correlation between MET receptor expression, MET gene alterations and the two most frequent occurring EGFR gene mutations was also investigated. Materials and methods Stage I to IIIA tumors from 326 patients with NSCLC were immunohistochemically tested for protein expression of MET and IGF-1. Their cytoplasmic expression was compared with the gene copy number of the MET and IGF1Rgenes by SISH in paraffin-embedded, formalin-fixed material. Correlations were made with the immunohistochemical expression of two frequent EGFR mutations and clinicopathological variables. Univariate and multivariate survival analyses was used to evaluate the prognostic efficacy of the tested markers. Results In univariate analyses, high cytoplasmic MET expression showed a significant negative prognostic effect in adenocarcinoma patients (p = 0.026). MET gene to chromosome 7 ratio was a significant positive prognostic marker (p = 0.005), probably only due to the highly negative prognostic significance of chromosome 7 polysomy (p = 0.002). High IGF1R gene copy number was a negative prognostic marker for all NSCLC patients (p = 0.037). In the multivariate analysis, polysomy of chromosome 7 in tumor cells correlated significantly and independently with a poor prognosis (p = 0.011). In patients with adenocarcinoma, a high cytoplasmic MET expression was an independent negative prognostic marker (p = 0.013). In males a high IGF1R gene copy number to chromosome 15 count ratio was significantly and independently correlated to a poor prognosis (p = 0.018). Conclusion MET protein expression provides superior prognostic information compared with SISH. Polysomy of chromosome 7 is an independent negative prognostic factor in NSCLC patients. This finding has an important implication while examining genes located on chromosome 7 by means of SISH. High IGF1R gene copy number to chromosome 15 count ratio is an independent predictor of inferior survival in male patients with primary NSCLC.
Collapse
Affiliation(s)
- Samer Al-Saad
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
- * E-mail:
| | - Elin Richardsen
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
| | - Thomas K. Kilvaer
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Tom Donnem
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Sigve Andersen
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Mehrdad Khanehkenari
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Roy M. Bremnes
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Lill-Tove Busund
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
| |
Collapse
|
214
|
Prevalence and clinical association of MET gene overexpression and amplification in patients with NSCLC: Results from the European Thoracic Oncology Platform (ETOP) Lungscape project. Lung Cancer 2017; 111:143-149. [PMID: 28838386 DOI: 10.1016/j.lungcan.2017.07.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/13/2017] [Accepted: 07/16/2017] [Indexed: 12/16/2022]
Abstract
INTRODUCTION In a well-defined NSCLC cohort of the ETOP Lungscape program, we explored the epidemiology of IHC MET overexpression and amplification, their inter-correlation, and their association to outcome. METHODS Resected NSCLC were assessed for MET gene copy number (GCN) and expression using silver in-situ hybridization (SISH) and immunohistochemistry (IHC) on TMAs in a multicenter setting. MET amplification was defined as MET/centromere ratio≥2 (with average MET GCN≥4), high MET GCN as CGN≥5 and MET IHC+ as ≥2+ intensity in ≥50% of tumor cells. A total of 182 MET IHC+ and EGFR/KRAS WT tumors were analyzed for METex14 skipping mutation. RESULTS MET IHC+ was found in 23.8% of 2432 patients, significantly associated with female gender, small tumor size, and adenocarcinoma histology. We observed a high inter-laboratory variability in IHC and SISH analysis. MET amplification prevailed in 4.6% and MET GCN≥5 in 4.1% of 1572 patients. MET amplification and MET GCN≥5 were not significantly associated with any tumor characteristics or stage. Both were significantly associated with IHC MET positivity (p<0.001). METex14 skipping mutation prevailed in 5 of 182 (2.7%) MET IHC+ WT EGFR/KRAS NSCLC, 4 of which within the 88 adenocarcinomas (4.5%). No association of IHC MET overexpression, SISH MET amplification or high MET GCN was found with OS, RFS or TTR. CONCLUSION MET overexpression is found in 23.8% of surgically resected NSCLC. MET amplification prevails in 4.6% and is associated with MET overexpression. Both have no influence on prognosis. The large inter-laboratory variability in IHC highlights the challenge of MET IHC analysis in routine practice.
Collapse
|
215
|
Preferential Localization of MET Expression at the Invasion Front and in Spreading Cells Through Air Spaces in Non-Small Cell Lung Carcinomas. Am J Surg Pathol 2017; 41:414-422. [PMID: 28098570 DOI: 10.1097/pas.0000000000000810] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The involvement of the HGF/MET pathway in acquisition of an invasive phenotype in non-small cell lung carcinomas (NSCLCs) suggests that MET inhibitors might prove effective against these cancers, but clinical trials have yielded conflicting results. The aim of our study was to evaluate how intratumoral heterogeneity (ITH) of MET staining affects the determination of MET status for therapeutic purposes. We analyzed 64 NSCLC samples, including 33 adenocarcinomas (ADCs) and 31 squamous cell carcinomas (SCCs). We used immunohistochemistry to detect MET and phospho-MET on whole slides and determined the MET SP44 immunoscore and the H-score. A high METMab score (2+/3+) was observed in 34% of NSCLCs and was more prevalent in ADCs (52%) than in SCCs (16%). We found ITH in 73% of ADCs and 77% of SCCs, with higher levels of MET and phospho-MET at the invasion front (in 52% of ADCs and 22% of SCCs) and in tumor cells spreading through air spaces in ADCs. Within-sample ITH was high in 40% of the ADCs and 29% of the SCCs. When different samples from the same tumor were compared, discordant assessments (high MET vs. low MET) were made for 12% of the ADCs and 10% of the SCCs. C-MET and phospho-MET overexpression occurred preferentially in ADCs and in areas involved in tumor progression, in support of the view that MET activation plays a role in the development of an invasive phenotype in NSCLC. To use MET status adequately as a biomarker, one must take the resulting high level of ITH into account.
Collapse
|
216
|
Higgins GS, Krause M, McKenna WG, Baumann M. Personalized Radiation Oncology: Epidermal Growth Factor Receptor and Other Receptor Tyrosine Kinase Inhibitors. Recent Results Cancer Res 2017; 198:107-22. [PMID: 27318683 DOI: 10.1007/978-3-662-49651-0_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Molecular biomarkers are currently evaluated in preclinical and clinical studies in order to establish predictors for treatment decisions in radiation oncology. The receptor tyrosine kinases (RTK) are described in the following text. Among them, the most data are available for the epidermal growth factor receptor (EGFR) that plays a major role for prognosis of patients after radiotherapy, but seems also to be involved in mechanisms of radioresistance, specifically in repopulation of tumour cells between radiotherapy fractions. Monoclonal antibodies against the EGFR improve locoregional tumour control and survival when applied during radiotherapy, however, the effects are heterogeneous and biomarkers for patient selection are warranted. Also other RTK´s such as c-Met and IGF-1R seem to play important roles in tumour radioresistance. Beside the potential to select patients for molecular targeting approaches combined with radiotherapy, studies are also needed to evluate radiotherapy adaptation approaches for selected patients, i.e. adaptation of radiation dose, or, more sophisticated, of target volumes.
Collapse
Affiliation(s)
- Geoff S Higgins
- Gray Laboratories, Department of Oncology, Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- German Cancer Consortium (DKTK) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Insititute of Radiooncology, Dresden, Germany.
- Department of Radiation Oncology, Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - W Gillies McKenna
- Gray Laboratories, Department of Oncology, Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Insititute of Radiooncology, Dresden, Germany
- Department of Radiation Oncology, Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
217
|
Poulsen TT, Grandal MM, Skartved NJØ, Hald R, Alifrangis L, Koefoed K, Lindsted T, Fröhlich C, Pollmann SE, Eriksen KW, Dahlman A, Jacobsen HJ, Bouquin T, Pedersen MW, Horak ID, Lantto J, Kragh M. Sym015: A Highly Efficacious Antibody Mixture against MET-Amplified Tumors. Clin Cancer Res 2017; 23:5923-5935. [PMID: 28679766 DOI: 10.1158/1078-0432.ccr-17-0782] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/01/2017] [Accepted: 06/29/2017] [Indexed: 02/03/2023]
Abstract
Purpose: Activation of the receptor tyrosine kinase MET is associated with poor clinical outcome in certain cancers. To target MET more effectively, we developed an antagonistic antibody mixture, Sym015, consisting of two humanized mAbs directed against nonoverlapping epitopes of MET.Experimental Design/Results: We screened a large panel of well-annotated human cancer cell lines and identified a subset with highly elevated MET expression. In particular, cell lines of lung cancer and gastric cancer origin demonstrated high MET expression and activation, and Sym015 triggered degradation of MET and significantly inhibited growth of these cell lines. Next, we tested Sym015 in patient- and cell line-derived xenograft models with high MET expression and/or MET exon 14 skipping alterations, and in models harboring MET amplification as a mechanism of resistance to EGFR-targeting agents. Sym015 effectively inhibited tumor growth in all these models and was superior to an analogue of emibetuzumab, a monoclonal IgG4 antibody against MET currently in clinical development. Sym015 also induced antibody-dependent cellular cytotoxicity (ADCC) in vitro, suggesting that secondary effector functions contribute to the efficacy of Sym015.Retrospectively, all responsive, high MET-expressing models were scored as highly MET-amplified by in situ hybridization, pointing to MET amplification as a predictive biomarker for efficacy. Preclinical toxicology studies in monkeys showed that Sym015 was well tolerated, with a pharmacokinetic profile supporting administration of Sym015 every second or third week in humans.Conclusions: The preclinical efficacy and safety data provide a clear rationale for the ongoing clinical studies of Sym015 in patients with MET-amplified tumors. Clin Cancer Res; 23(19); 5923-35. ©2017 AACR.
Collapse
|
218
|
Tang Q, Wang L, Duan Y, Wang W, Huang S, Zhi J, Jia S, Zhu W, Wang P, Luo R, Zheng P. Discovery of novel 7-azaindole derivatives bearing dihydropyridazine moiety as c-Met kinase inhibitors. Eur J Med Chem 2017; 133:97-106. [DOI: 10.1016/j.ejmech.2017.03.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/22/2017] [Indexed: 01/20/2023]
|
219
|
Pei J, Chu T, Shao M, Teng J, Sha H, Gu A, Li R, Qian J, Mao W, Li Y, Han B. Potential Antitumor Activity of SIM-89 in Non-Small Cell Lung Cancer Cells. Yonsei Med J 2017; 58:581-591. [PMID: 28332364 PMCID: PMC5368144 DOI: 10.3349/ymj.2017.58.3.581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 11/27/2022] Open
Abstract
PURPOSE c-Met and its ligand, hepatocyte growth factor (HGF), play a critical role in oncogenesis and metastatic progression. The aim of this study was to identify inhibited enzymogram and to test the antitumor activity of SIM-89 (a c-Met receptor tyrosine kinase inhibitor) in non-small cell lung cancer. MATERIALS AND METHODS Z'-LYTE kinase assay was employed to screen the kinase enzymogram, and mechanism of action (MOA) analysis was used to identify the inhibited kinases. Cell proliferation was then analyzed by CCK8 assay, and cell migration was determined by transwell assay. The gene expression and the phosphorylation of c-Met were examined by realtime-PCR and western blotting, respectively. Finally, the secretion of HGF was detected by ELISA assay. RESULTS c-Met, activated protein kinase (AMPK), and tyrosine kinase A (TRKA) were inhibited by SIM-89 with the IC₅₀ values of 297 nmol/L, 1.31 μmol/L, and 150.2 nmol/L, respectively. SIM-89 exerted adenosine triphosphate (ATP) competitive inhibition on c-Met. Moreover, the expressions of STAT1, JAK1, and c-Met in H460 cells were decreased by SIM-89 treatment, and c-Met phosphorylation was suppressed in A549, H441, H1299, and B16F10 cells by the treatment. In addition, SIM-89 treatment significantly decreased the level of HGF, which accounted for the activation of c-Met receptor tyrosine kinase. Finally, we showed cell proliferation inhibition and cell migration suppression in H460 and H1299 cells after SIM-89 treatment. CONCLUSION In conclusion, SIM-89 inhibits tumor cell proliferation, migration and HGF autocrine, suggesting it's potential antitumor activity.
Collapse
Affiliation(s)
- Jun Pei
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tianqing Chu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Minhua Shao
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiajun Teng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huifang Sha
- Department of Basic Research, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Aiqing Gu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Li
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jialin Qian
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weifeng Mao
- Department of Biology, East China Normal University, Shanghai, China
| | - Ying Li
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
220
|
MET exon 14 skipping mutation in triple-negative pulmonary adenocarcinomas and pleomorphic carcinomas: An analysis of intratumoral MET status heterogeneity and clinicopathological characteristics. Lung Cancer 2017; 106:131-137. [DOI: 10.1016/j.lungcan.2017.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/13/2016] [Accepted: 02/10/2017] [Indexed: 11/15/2022]
|
221
|
Friese-Hamim M, Bladt F, Locatelli G, Stammberger U, Blaukat A. The selective c-Met inhibitor tepotinib can overcome epidermal growth factor receptor inhibitor resistance mediated by aberrant c-Met activation in NSCLC models. Am J Cancer Res 2017; 7:962-972. [PMID: 28469968 PMCID: PMC5411803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/27/2017] [Indexed: 06/07/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) sensitive to first-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) often acquires resistance through secondary EGFR mutations, including the T790M mutation, aberrant c-Met receptor activity, or both. We assessed the ability of the highly selective c-Met inhibitor tepotinib to overcome EGFR TKI resistance in various xenograft models of NSCLC. In models with EGFR-activating mutations and low c-Met expression (patient explant-derived LU342, cell line PC-9), EGFR TKIs caused tumors to shrink, but growth resumed upon cessation of treatment. Tepotinib combined with EGFR TKIs delayed tumor regrowth, while tepotinib alone was ineffective. In patient explant-derived LU858, which has an EGFR-activating mutation and expresses high levels of c-Met/HGF, EGFR TKIs had no effect on tumor growth. Tepotinib combined with EGFR TKIs caused complete tumor regression and tepotinib alone caused tumor stasis. In cell line DFCI081 (activating EGFR mutation, c-Met amplification), EGFR TKIs were ineffective, whereas tepotinib alone induced complete tumor regression. Finally, in a 'double resistant' EGFR T790M-positive, high c-Met model (cell line HCC827-GR-T790M), the EGFR TKIs erlotinib, afatinib, and rociletinib, as well as tepotinib as a single agent or in combination with erlotinib or afatinib, slowed tumor growth, but only tepotinib in combination with rociletinib induced complete tumor regression. We conclude that tepotinib can overcome acquired resistance to EGFR TKIs. Based on these data, clinical trials of tepotinib in combination with EGFR TKIs in patients with NSCLC with acquired resistance to first-generation EGFR TKIs are warranted.
Collapse
Affiliation(s)
| | - Friedhelm Bladt
- Translational and Biomarker Research, Merck KGaADarmstadt, Germany
| | | | - Uz Stammberger
- Global Research and Development, Merck KGaADarmstadt, Germany
| | - Andree Blaukat
- Global Research and Development, Merck KGaADarmstadt, Germany
| |
Collapse
|
222
|
Ortiz-Zapater E, Lee RW, Owen W, Weitsman G, Fruhwirth G, Dunn RG, Neat MJ, McCaughan F, Parker P, Ng T, Santis G. MET-EGFR dimerization in lung adenocarcinoma is dependent on EGFR mtations and altered by MET kinase inhibition. PLoS One 2017; 12:e0170798. [PMID: 28141869 PMCID: PMC5283661 DOI: 10.1371/journal.pone.0170798] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/11/2017] [Indexed: 01/06/2023] Open
Abstract
Advanced lung cancer has poor survival with few therapies. EGFR tyrosine kinase inhibitors (TKIs) have high response rates in patients with activating EGFR mutations, but acquired resistance is inevitable. Acquisition of the EGFR T790M mutation causes over 50% of resistance; MET amplification is also common. Preclinical data suggest synergy between MET and EGFR inhibitors. We hypothesized that EGFR-MET dimerization determines response to MET inhibition, depending on EGFR mutation status, independently of MET copy number. We tested this hypothesis by generating isogenic cell lines from NCI-H1975 cells, which co-express L858R and T790M EGFR mutations, namely H1975L858R/T790M (EGFR TKI resistant); H1975L858R (sensitized) and H1975WT (wild-type). We assessed cell proliferation in vitro and tumor growth/stroma formation in derived xenograft models in response to a MET TKI (SGX523) and correlated with EGFR-MET dimerization assessed by Förster Resonance Energy Transfer (FRET). SGX523 significantly reduced H1975L858R/T790M cell proliferation, xenograft tumor growth and decreased ERK phosphorylation. The same was not seen in H1975L858R or H1975WT cells. SGX523 only reduced stroma formation in H1975L858R. SGX523 reduced EGFR-MET dimerization in H1975L858R/T790M but induced dimer formation in H1975L858R with no effect in H1975WT. Our data suggests that MET inhibition by SGX523 and EGFR-MET heterodimerisation are determined by EGFR genotype. As tumor behaviour is modulated by this interaction, this could determine treatment efficacy.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Division of Asthma, Allergy and Lung Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Richard W. Lee
- Division of Asthma, Allergy and Lung Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - William Owen
- Division of Asthma, Allergy and Lung Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Gregory Weitsman
- Richard Dimbleby Department of Cancer Research, Randall Division of Cell and Molecular Biophysics, King’s College London, Guy's Medical School Campus, London, United Kingdom
| | - Gilbert Fruhwirth
- Department of Imaging Chemistry and Biology, Division of Imaging Science and Biomedical Engineering, King’s College London, The Rayne Institute/St. Thomas' Hospital, London, United Kingdom
| | - Robert G. Dunn
- Department of Cancer Genetics, Viapath, Guy’s and St Thomas’ NHS Foundation Trust, Guy's Hospital, London, United Kingdom
| | - Michael J. Neat
- Department of Cancer Genetics, Viapath, Guy’s and St Thomas’ NHS Foundation Trust, Guy's Hospital, London, United Kingdom
| | - Frank McCaughan
- Division of Asthma, Allergy and Lung Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Peter Parker
- Division of Cancer Studies, King’s College London, Guy's Medical School Campus, London, United Kingdom
- Protein Phosphorylation Laboratory, Francis Crick Institute, London, United Kingdom
| | - Tony Ng
- Richard Dimbleby Department of Cancer Research, Randall Division of Cell and Molecular Biophysics, King’s College London, Guy's Medical School Campus, London, United Kingdom
- Division of Cancer Studies, King’s College London, Guy's Medical School Campus, London, United Kingdom
- Breast Cancer Research Unit, King’s College London, Guy's Hospital, London, United Kingdom
- UCL Cancer Institute, Paul O' Gorman Building, University College London, London, United Kingdom
| | - George Santis
- Division of Asthma, Allergy and Lung Biology, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
223
|
Kwon MJ, Kim JW, Jeon JY, Nam ES, Cho SJ, Park HR, Min SK, Seo J, Min KW, Choe JY, Lee HK. Concurrent MET copy number gain and KRAS mutation is a poor prognostic factor in pancreatobiliary subtype ampullary cancers. Pathol Res Pract 2017; 213:381-388. [PMID: 28214200 DOI: 10.1016/j.prp.2017.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/16/2016] [Accepted: 01/06/2017] [Indexed: 12/18/2022]
Abstract
Hepatocyte growth factor (HGF) and MET are candidates of targeted therapies for cancer patients. Although MET and HGF are commonly expressed in biliary tract cancers, their expression and gene copy number status and their association with KRAS mutations have not been investigated in pancreatobiliary-type ampullary adenocarcinomas (A-ACs), one of the aggressive periampullary cancers. MET and HGF expressions and MET copy number status were examined by performing immunohistochemistry (IHC) and silver in situ hybridization (SISH) in 62 surgically resected, paraffin-embedded tumors, respectively. High MET and HGF protein expressions were detected in 24 (38.7%) and 15 (24.2%) tumors. High MET expression was associated with KRAS mutation. However, there were no associations of high MET/HGF expression alone with other clinicopathological feature or survival. MET SISH positivity was detected in 19 tumors (30.6%), where 84.2% were due to high trisomy or polysomy and only 3 cases (15.8%) were MET gene amplification. The overall MET protein overexpression was well correlated with MET SISH positivity. The concurrent MET SISH positivity and KRAS mutation, not each alone, was an independent poor prognostic factor of disease-free survival only in pancreatobiliary subtype of A-ACs, but not in intestinal subtype. Concurrent MET SISH positivity and KRAS mutation may predict a high risk of recurrence in pancreatobiliary subtype of A-ACs, indicating those markers could be potent candidates for a new therapeutic target in this cancer type. MET IHC can be used as a reliable tool screening for MET copy number status in ampullary cancers.
Collapse
Affiliation(s)
- Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 431-070, Republic of Korea.
| | - Jeong Won Kim
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Daerim 1-Dong, Yeongdeungpo-gu, Seoul 150-950, Republic of Korea
| | - Jang Yong Jeon
- Division of Hepato-Billiary-Pancreatic Surgery and Liver Transplantation, Department of Surgery, Hangang Sacred Heart Hospital, Hallym University College of Medicine, 12, Beodeunaru-ro 7-gil, Yeongdeungpo-gu, Seoul 150-719, Republic of Korea
| | - Eun Sook Nam
- Department of Pathology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 134-701, Republic of Korea
| | - Seong Jin Cho
- Department of Pathology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 134-701, Republic of Korea
| | - Hye-Rim Park
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 431-070, Republic of Korea
| | - Soo Kee Min
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 431-070, Republic of Korea
| | - Jinwon Seo
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 431-070, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Kyoungchun-ro 153, Guri-si, Gyeonggi-do 11923, Republic of Korea
| | - Ji-Young Choe
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 431-070, Republic of Korea
| | - Hye Kyung Lee
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 431-070, Republic of Korea
| |
Collapse
|
224
|
Wakelee H, Zvirbule Z, De Braud F, Kingsley CD, Mekhail T, Lowe T, Schütte W, Lena H, Lawler W, Braiteh F, Cosgriff T, Kaen D, Boyer M, Hsu J, Phan S, Novello S. Efficacy and Safety of Onartuzumab in Combination With First-Line Bevacizumab- or Pemetrexed-Based Chemotherapy Regimens in Advanced Non-Squamous Non–Small-Cell Lung Cancer. Clin Lung Cancer 2017; 18:50-59. [DOI: 10.1016/j.cllc.2016.09.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/31/2016] [Accepted: 09/06/2016] [Indexed: 01/24/2023]
|
225
|
Drilon A, Cappuzzo F, Ou SHI, Camidge DR. Targeting MET in Lung Cancer: Will Expectations Finally Be MET? J Thorac Oncol 2017; 12:15-26. [PMID: 27794501 PMCID: PMC5603268 DOI: 10.1016/j.jtho.2016.10.014] [Citation(s) in RCA: 290] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/12/2016] [Accepted: 10/16/2016] [Indexed: 12/12/2022]
Abstract
The hepatocyte growth factor receptor (MET) is a potential therapeutic target in a number of cancers, including NSCLC. In NSCLC, MET pathway activation is thought to occur through a diverse set of mechanisms that influence properties affecting cancer cell survival, growth, and invasiveness. Preclinical and clinical evidence suggests a role for MET activation as both a primary oncogenic driver in subsets of lung cancer and as a secondary driver of acquired resistance to targeted therapy in other genomic subsets. In this review, we explore the biology and clinical significance behind MET proto-oncogene receptor tyrosine kinase (MET) exon 14 alterations and MET amplification in NSCLC, the role of MET amplification in the setting of acquired resistance to EGFR tyrosine kinase inhibitor therapy in EGFR-mutant NSCLC, and the history of MET pathway inhibitor drug development in NSCLC, highlighting current strategies that enrich for biomarkers likely to be predictive of response. Whereas previous trials that focused on MET pathway-directed targeted therapy in unselected or MET-overexpressing NSCLC yielded largely negative results, more recent investigations focusing on MET exon 14 alterations and MET amplification have been notable for meaningful clinical responses to MET inhibitor therapy in a substantial proportion of patients.
Collapse
Affiliation(s)
| | | | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California
| | | |
Collapse
|
226
|
İşcan E, Güneş A, Korhan P, Yılmaz Y, Erdal E, Atabey N. The regulatory role of heparin on c-Met signaling in hepatocellular carcinoma cells. J Cell Commun Signal 2016; 11:155-166. [PMID: 27975162 DOI: 10.1007/s12079-016-0368-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
The role of heparin as an anticoagulant is well defined; however, its role in tumorigenesis and tumor progression is not clear yet. Some studies have shown that anticoagulant treatment in cancer patients improve overall survival, however, recent clinical trials have not shown a survival benefit in cancer patients receiving heparin treatment. In our previous studies we have shown the inhibitory effects of heparin on Hepatocyte Growth Factor (HGF)-induced invasion and migration in hepatocellular carcinoma (HCC) cells. In this study, we showed the differential effects of heparin on the behaviors of HCC cells based on the presence or absence of HGF. In the absence of HGF, heparin activated HGF/c-Met signaling and promoted motility and invasion in HCC cells. Heparin treatment led to c-Met receptor dimerization and activated c-Met signaling in an HGF independent manner. Heparin-induced c-Met activation increased migration and invasion through ERK1/2, early growth response factor 1 (EGR1) and Matrix Metalloproteinases (MMP) axis. Interestingly, heparin modestly decreased the proliferation of HCC cells by inhibiting activatory phosphorylation of Akt. The inhibition of c-Met signaling reversed heparin-induced increase in motility and invasion and, proliferation inhibition. Our study provides a new perspective into the role of heparin on c-Met signaling in HCC.
Collapse
Affiliation(s)
- Evin İşcan
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balçova, Izmir, Turkey
| | - Aysim Güneş
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balçova, Izmir, Turkey.,Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Peyda Korhan
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Yeliz Yılmaz
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balçova, Izmir, Turkey.,Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Esra Erdal
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balçova, Izmir, Turkey.,Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Neşe Atabey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balçova, Izmir, Turkey. .,Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey.
| |
Collapse
|
227
|
Spigel DR, Edelman MJ, O'Byrne K, Paz-Ares L, Mocci S, Phan S, Shames DS, Smith D, Yu W, Paton VE, Mok T. Results From the Phase III Randomized Trial of Onartuzumab Plus Erlotinib Versus Erlotinib in Previously Treated Stage IIIB or IV Non-Small-Cell Lung Cancer: METLung. J Clin Oncol 2016; 35:412-420. [PMID: 27937096 DOI: 10.1200/jco.2016.69.2160] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose The phase III OAM4971g study (METLung) examined the efficacy and safety of onartuzumab plus erlotinib in patients with locally advanced or metastatic non-small-cell lung cancer selected by MET immunohistochemistry whose disease had progressed after treatment with a platinum-based chemotherapy regimen. Patients and Methods Patients were randomly assigned at a one-to-one ratio to receive onartuzumab (15 mg/kg intravenously on day 1 of each 21-day cycle) plus daily oral erlotinib 150 mg or intravenous placebo plus daily oral erlotinib 150 mg. The primary end point was overall survival (OS) in the intent-to-treat population. Secondary end points included median progression-free survival, overall response rate, biomarker analysis, and safety. Results A total of 499 patients were enrolled (onartuzumab, n = 250; placebo, n = 249). Median OS was 6.8 versus 9.1 months for onartuzumab versus placebo (stratified hazard ratio [HR], 1.27; 95% CI, 0.98 to 1.65; P = .067), with a greater number of deaths in the onartuzumab arm (130 [52%] v 114 [46%]). Median progression-free survival was 2.7 versus 2.6 months (stratified HR, 0.99; 95% CI, 0.81 to 1.20; P = .92), and overall response rate was 8.4% and 9.6% for onartuzumab versus placebo, respectively. Exploratory analyses using MET fluorescence in situ hybridization status and gene expression showed no benefit for onartuzumab; patients with EGFR mutations showed a trend toward shorter OS with onartuzumab treatment (HR, 4.68; 95% CI, 0.97 to 22.63). Grade 3 to 5 adverse events were reported by 56.0% and 51.2% of patients, with serious AEs in 33.9% and 30.7%, for experimental versus control arms, respectively. Conclusion Onartuzumab plus erlotinib did not improve clinical outcomes, with shorter OS in the onartuzumab arm, compared with erlotinib in patients with MET-positive non-small-cell lung cancer.
Collapse
Affiliation(s)
- David R Spigel
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Martin J Edelman
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Kenneth O'Byrne
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Luis Paz-Ares
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Simonetta Mocci
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - See Phan
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - David S Shames
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Dustin Smith
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Wei Yu
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Virginia E Paton
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Tony Mok
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| |
Collapse
|
228
|
Catenacci DVT, Ang A, Liao WL, Shen J, O'Day E, Loberg RD, Cecchi F, Hembrough T, Ruzzo A, Graziano F. MET tyrosine kinase receptor expression and amplification as prognostic biomarkers of survival in gastroesophageal adenocarcinoma. Cancer 2016; 123:1061-1070. [PMID: 27926778 PMCID: PMC5339041 DOI: 10.1002/cncr.30437] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/14/2016] [Accepted: 10/05/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND MET gene amplification and Met protein overexpression may be associated with a poor prognosis. The MET/Met status is typically determined with fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC), respectively. Targeted proteomics uses mass spectrometry–based selected reaction monitoring (SRM) to accurately quantitate Met expression. FISH, IHC, and SRM analyses were compared to characterize the prognostic value of MET/Met in gastroesophageal adenocarcinoma (GEC). METHODS Samples from 447 GEC patients were analyzed for MET gene amplification (FISH) and Met protein expression (IHC and SRM). Cox proportional hazards models and Kaplan‐Meier estimates were applied to explore relations between Met, overall survival (OS), and clinical/pathological characteristics. Spearman's rank coefficient was used to assess the correlation between parameters. RESULTS Patients with MET‐amplified tumors had worse OS when: the MET/centromere enumeration probe for chromosome 7 FISH ratio was ≥ 2 (hazard ratio [HR], 3.13; 95% confidence interval [CI], 1.84‐5.33), the MET gene copy number was ≥5 (HR, 2.51; 95% CI, 1.45‐4.34), or ≥ 10% of the cells had ≥15 copies (HR, 4.28; 95% CI, 2.18‐8.39). Similar observations were made with Met protein overexpression by IHC (≥1 + intensity in ≥ 25% of the tumor cell membrane: HR, 1.39; 95% CI, 1.04‐1.86) or SRM (≥400 amol/μg: HR, 1.76; 95% CI, 1.06‐2.90). A significant correlation was observed between MET FISH/Met IHC, MET FISH/Met SRM, and Met IHC/Met SRM; only MET FISH and Met SRM were independent negative prognostic biomarkers in multivariate analyses. CONCLUSIONS MET amplification and overexpression, assessed by multiple methods, were associated with a worse prognosis in univariate analyses. However, only MET amplification by FISH and Met expression by SRM were independent prognostic biomarkers. Compared with IHC, SRM may provide an added benefit for informed decisions about Met‐targeted therapy. Cancer 2017;123:1061–70. © 2016 American Cancer Society. In a large study, MET gene amplification, Met protein overexpression, or both, as assessed by various assays, are associated with a poor prognosis in univariate analyses. However, only MET amplification by fluorescence in situ hybridization and Met expression by selected reaction monitoring mass spectrometry are independent prognostic biomarkers; compared with immunohistochemistry, selected reaction monitoring may provide an added benefit for informed decisions about Met‐targeted therapy.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Agnes Ang
- Amgen, Inc, Thousand Oaks, California
| | | | - Jing Shen
- Amgen, Inc, Thousand Oaks, California
| | - Emily O'Day
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | | | | | | | - Annamaria Ruzzo
- Department of Biomolecular Science, University of Urbino, Urbino, Italy
| | - Francesco Graziano
- Department of Onco-Hematology, Azienda Ospedali Riuniti Marche Nord, Pesaro, Italy
| |
Collapse
|
229
|
Michels S, Wolf J. Stratified Treatment in Lung Cancer. Oncol Res Treat 2016; 39:760-766. [PMID: 27889787 DOI: 10.1159/000453406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
Even though great efforts have been made to improve chemotherapy-based treatment approaches for lung cancer, the prognosis of patients with advanced and metastasized disease remains particularly poor. In recent years, a growing number of genetic aberrations driving lung cancer have been identified. Targeted inhibition of some of these aberrations, most prominently mutated EGFR and ALK, by tyrosine kinase inhibitors has dramatically increased efficacy and tolerability of systemic lung cancer treatment in subsets of patients. However, the duration of response is limited due to the acquisition of molecular mechanisms of resistance to targeted treatment. Modern next-generation inhibitors aim to break resistance. A deep understanding of the mechanisms of treatment failure is imperative to the development of new approaches. In this review, we focus on the current status of stratified therapy in lung cancer and highlight new, potentially promising treatment approaches.
Collapse
Affiliation(s)
- Sebastian Michels
- Lung Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | | |
Collapse
|
230
|
Reungwetwattana T, Liang Y, Zhu V, Ou SHI. The race to target MET exon 14 skipping alterations in non-small cell lung cancer: The Why, the How, the Who, the Unknown, and the Inevitable. Lung Cancer 2016; 103:27-37. [PMID: 28024693 DOI: 10.1016/j.lungcan.2016.11.011] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/11/2016] [Accepted: 11/13/2016] [Indexed: 01/29/2023]
Abstract
A number of small molecule tyrosine kinase inhibitors (TKIs) have now been approved for the treatment of non-small cell lung cancers (NSCLC), including those targeted against epidermal growth factor receptor, anaplastic lymphoma kinase, and ROS1. Despite a wealth of agents developed to target the receptor tyrosine kinase, MET, clinical outcomes have as yet been disappointing, leading to pessimism about the role of MET in the pathogenesis of NSCLC. However, in recent years, there has been a renewed interest in MET exon 14 alterations as potential drivers of lung cancer. MET exon 14 alterations, which result in increased MET protein levels due to disrupted ubiquitin-mediated degradation, occur at a prevalence of around 3% in adenocarcinomas and around 2% in other lung neoplasms, making them attractive targets for the treatment of lung cancer. At least five MET-targeted TKIs, including crizotinib, cabozantinib, capmatinib, tepotinib, and glesatinib, are being investigated clinically for patients with MET exon 14 altered-NSCLC. A further two compounds have shown activity in preclinical models. In this article, we review the current clinical and preclinical data available for these TKIs, along with a number of other potential therapeutic options, including antibodies and immunotherapy. A number of questions remain unanswered regarding the future of MET TKIs, but unfortunately, the development of resistance to targeted therapies is inevitable. Resistance is expected to arise as a result of receptor tyrosine kinase mutation or from upregulation of MET ligand expression; potential strategies to overcome resistance are proposed.
Collapse
Affiliation(s)
- Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Ying Liang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Viola Zhu
- Long Beach Veterans Administration Hospital, Long Beach, CA 90822, USA; Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange County, CA 92868, USA
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange County, CA 92868, USA.
| |
Collapse
|
231
|
Yuen HF, Chan KK, Platt-Higgins A, Dakir EH, Matchett KB, Haggag YA, Jithesh PV, Habib T, Faheem A, Dean FA, Morgan R, Rudland PS, El-Tanani M. Ran GTPase promotes cancer progression via Met recepto-rmediated downstream signaling. Oncotarget 2016; 7:75854-75864. [PMID: 27716616 PMCID: PMC5342783 DOI: 10.18632/oncotarget.12420] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/21/2016] [Indexed: 01/12/2023] Open
Abstract
It has been shown previously that cancer cells with an activated oncogenic pathway, including Met activation, require Ran for growth and survival.Here, we show that knockdown of Ran leads to a reduction of Met receptor expression in several breast and lung cancer cell lines. This, in turn suppressed HGF expression and the Met-mediated activation of the Akt pathway, as well as cell adhesion, migration, and invasion. In a cell line model where Met amplification has previously been shown to contribute to gefitinib resistance, Ran knockdown sensitized cells to gefitinib-mediated inhibition of Akt and ERK1/2 phosphorylation and consequently reduced cell proliferation. We further demonstrate that Met reduction-mediated by knockdown of Ran, occurs at the post-transcriptional level, probably via a matrix metalloproteinase. Moreover, the level of immunoreactive Ran and Met are positively associated in human breast cancer specimens, suggesting that a high level of Ran may be a pre-requisite for Met overexpression. Interestingly, a high level of immunoreactive Ran dictates the prognostic significance of Met, indicating that the co-overexpression of Met and Ran may be associated with cancer progression and could be used in combination as a prognostic indicator.
Collapse
Affiliation(s)
- Hiu-Fung Yuen
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Ka-Kui Chan
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Angela Platt-Higgins
- Cancer and Polio Research Fund Laboratories, School of Biological Sciences, University of Liverpool, Liverpool, UK
| | - El-Habib Dakir
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
- Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire, UK
| | - Kyle B. Matchett
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Yusuf Ahmed Haggag
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Tanta, Tanta, Egypt
| | - Puthen V. Jithesh
- Biomedical Informatics Research, Sidra Medical and Research Center, Doha, Qatar
| | - Tanwir Habib
- Biomedical Informatics Research, Sidra Medical and Research Center, Doha, Qatar
| | - Ahmed Faheem
- University of Sunderland, Department of Pharmacy, Health and Well-Being, Sunderland Pharmacy School, Sunderland, UK
| | - Fennell A. Dean
- Translational Clinical Research, University of Leicester, Leicester, UK
| | - Richard Morgan
- Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire, UK
| | - Philip S. Rudland
- Cancer and Polio Research Fund Laboratories, School of Biological Sciences, University of Liverpool, Liverpool, UK
| | - Mohamed El-Tanani
- Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire, UK
| |
Collapse
|
232
|
Waxing and Waning of MET Amplification in EGFR-Mutated NSCLC in Response to the Presence and Absence of Erlotinib Selection Pressure. J Thorac Oncol 2016; 10:e115-8. [PMID: 26709484 DOI: 10.1097/jto.0000000000000642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
233
|
Cho YA, Kim EK, Heo SJ, Cho BC, Kim HR, Chung JM, Yoon SO. Alteration status and prognostic value of MET in head and neck squamous cell carcinoma. J Cancer 2016; 7:2197-2206. [PMID: 27994655 PMCID: PMC5166528 DOI: 10.7150/jca.16686] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 09/18/2016] [Indexed: 12/17/2022] Open
Abstract
The MET pathway plays a key role in various cancers, and its inhibition represents a potential treatment target. However, appropriate biomarkers are needed to facilitate the selection of patients who would benefit from MET inhibiting therapy. We herein conducted a robust confirmatory evaluation of the MET copy number alteration status and prognostic significance of c-Met expression in a large series of patients (n = 396) who underwent standard surgical resection and adjuvant chemoradiotherapy for head and neck squamous cell carcinoma (HNSCC). Surgically resected HNSCC samples were subjected to immunohistochemical and H-score analysis of c-Met expression and silver in situ hybridization analysis of MET amplification and copy number gains. c-Met expression varied, with mean and median H-scores (scale: 0-300 scale) of 61.2 and 60.0, respectively. The lowest and highest expression levels were observed in SCC of the larynx and oral cavity, respectively. MET copy number gains were observed in 16.9% of cases (67/339) and were associated with c-Met protein expression. High c-Met expression, determined according to MET gain status, was associated with an inferior overall survival rate, especially among completely resected cases. In conclusion, our robust analysis revealed that c-Met expression in HNSCCs varied according to anatomical site, correlated with MET copy number gains, and was associated with poor prognosis. This c-Met expression analysis method, which is based on the MET gain status, appears to appropriately predict high-risk HNSCC patients in the context of anti-MET therapeutic decisions.
Collapse
Affiliation(s)
- Yoon Ah Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Kyung Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Su Jin Heo
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Byoung Chul Cho
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Ryun Kim
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | | | - Sun Och Yoon
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
234
|
Kim IH, Lee IH, Lee JE, Hong SH, Kim TJ, Lee KY, Kim YK, Kim SJ, Sung SW, Park JK, Yoo IR, Kim YS, Kim JO, Kang JH. Clinical Significance of C-MET Overexpression and Epidermal Growth Factor Receptor Mutation in Platinum-Based Adjuvant Chemotherapy Outcome in Surgically Resected Lung Adenocarcinoma. Ann Surg Oncol 2016; 24:770-777. [PMID: 27696172 DOI: 10.1245/s10434-016-5599-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 11/18/2022]
Abstract
PURPOSE We retrospectively assessed the role of C-MET expression and epidermal growth factor receptor (EGFR) mutation on survival following platinum-based adjuvant chemotherapy. The impact of C-MET on survival was also investigated in relation to EGFR mutation status. METHODS We enrolled 311 patients with resected lung adenocarcinoma (high-risk stage 1B-3A), and performed immunohistochemistry (IHC) using C-MET- and mutant EGFR (EGFRmut)-specific antibodies in tissue microarrays. RESULTS Adjuvant chemotherapy was administered to 151 patients, 96 of whom relapsed and 85 died by the end of the study. On IHC, C-MET and EGFRmut were positive in 141 (45.3 %) and 88 (28.3 %) cases, respectively. On univariate analysis, adjuvant chemotherapy prolonged relapse-free survival (RFS) and overall survival (OS) in C-MET(+) patients (RFS p = 0.035; OS p = 0.013) but not in C-MET(-) patients. On multivariate analysis, adjuvant chemotherapy was a positive independent prognostic factor in C-MET(+) (RFS p = 0.013; OS p = 0.006) but not in C-MET(-) patients. In addition, univariate analysis showed no effect of EGFRmut status on RFS and OS after chemotherapy, whereas multivariate analysis revealed that adjuvant chemotherapy increased RFS in both EGFRmut(+) and EGFRmut(-) patients [EGFRmut(+) p = 0.033; EGFRmut(-) p = 0.030]. C-MET was a negative prognostic factor for RFS (p = 0.045) and OS (p = 0.007) in the EGFRmut(-) group but not in the EGFRmut(+) group, on multivariate analysis. CONCLUSIONS Our data indicate that patients with C-MET overexpression should be considered for adjuvant chemotherapy, and that C-MET negatively correlates with survival in patients with wild-type, but not mutant, EGFR.
Collapse
Affiliation(s)
- In-Ho Kim
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - In Hee Lee
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ji Eun Lee
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sook Hee Hong
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Tae-Jung Kim
- Department of Pathology, Yeouidol St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Kyo-Young Lee
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Young Kyoon Kim
- Department of Internal Medicine, Division of Pulmonology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Seung Joon Kim
- Department of Internal Medicine, Division of Pulmonology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sook Whan Sung
- Department of Thoracic Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jae Kil Park
- Department of Thoracic Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ie Ryung Yoo
- Department of Nuclear Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Yeon Sil Kim
- Department of Radiation Oncology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jung-Oh Kim
- Department of Biomedical Sciences, The Catholic University of Korea, Seoul, Korea
| | - Jin Hyoung Kang
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea. .,Multidisciplinary Team of Lung Cancer of Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
235
|
Li G, Gao Y, Cui Y, Zhang T, Cui R, Jiang Y, Shi J. Overexpression of CD44 is associated with the occurrence and migration of non-small cell lung cancer. Mol Med Rep 2016; 14:3159-67. [PMID: 27573351 PMCID: PMC5042784 DOI: 10.3892/mmr.2016.5636] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/07/2016] [Indexed: 11/17/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a potentially fatal disease and the incidence is increasing annually. In order to diagnose and treat NSCLC effectively, greater understanding of its molecular mechanism is required. In the present study, 36 NSCLC tissues and 10 normal tissues were selected. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to analyze the CD44 mRNA expression level in NSCLC tissue and DNA sequencing was performed to further verify the CD44 expression level. Differentially expressed genes between tumor tissues and controls were determined by DNA sequencing and the Gene_act_net between CD44 and its associated genes was constructed. Gene Ontology (GO) term enrichment analysis of the differentially expressed genes was performed by the Biological Networks Gene Ontology tool. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was performed based on the Expression Analysis Systematic Explorer test applied in the Database for Annotation, Visualization and Integrated Discovery. RT-qPCR results showed that CD34 was overexpressed in 21 of the 36 NSCLC tissues (58.3%). The Gene_act_net indicated that there were 20 differentially expressed genes with 17 upregulated and 3 downregulated. Among them, CD44, MET, ERBB2, EGFR, AKT1, IQGAP1 and STAT3 were associated with the occurrence and migration of NSCLC. In KEGG pathway analysis, extracellular matrix-receptor interaction and hematopoietic cell lineage pathways were the most affected by overexpressed CD44; and thus may be important in the development and migration of NSCLC. In conclusion, CD44 was overexpressed in NSCLC and the overexpression was associated with the occurrence of NSCLC and migration of NSCLC cells.
Collapse
Affiliation(s)
- Guanghu Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yufei Gao
- Department of Neurosurgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yongsheng Cui
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tao Zhang
- Department of Gastrointestinal Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Rui Cui
- Department of Laboratory Medicine Center, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yang Jiang
- Department of Colorectal Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Jingwei Shi
- Department of Laboratory Medicine Center, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
236
|
Yang T, Ng WH, Chen H, Chomchopbun K, Huynh TH, Go ML, Kon OL. Mitochondrial-Targeting MET Kinase Inhibitor Kills Erlotinib-Resistant Lung Cancer Cells. ACS Med Chem Lett 2016; 7:807-12. [PMID: 27563407 DOI: 10.1021/acsmedchemlett.6b00223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/23/2016] [Indexed: 12/13/2022] Open
Abstract
Lung cancer cells harboring activating EGFR mutations acquire resistance to EGFR tyrosine kinase inhibitors (TKIs) by activating several bypass mechanisms, including MET amplification and overexpression. We show that a significant proportion of activated MET protein in EGFR TKI-resistant HCC827 lung cancer cells resides within the mitochondria. Targeting the total complement of MET in the plasma membrane and mitochondria should render these cells more susceptible to cell death and hence provide a means of circumventing drug resistance. Herein, the mitochondrial targeting triphenylphosphonium (TPP) moiety was introduced to the selective MET kinase inhibitor PHA665752. The resulting TPP analogue rapidly localized to the mitochondria of MET-overexpressing erlotinib-resistant HCC827 cells, partially suppressed the phosphorylation (Y1234/Y1235) of MET in the mitochondrial inner membrane and was as cytotoxic and apoptogenic as the parent compound. These findings provide support for the targeting of mitochondrial MET with a TPP-TKI conjugate as a means of restoring responsiveness to chemotherapy.
Collapse
Affiliation(s)
- Tianming Yang
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | | | - Huan Chen
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Kamon Chomchopbun
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | | | - Mei Lin Go
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Oi Lian Kon
- Department
of Biochemistry, National University of Singapore, 8 Medical
Drive, Singapore 117596
| |
Collapse
|
237
|
Mok TSK, Geater SL, Su WC, Tan EH, Yang JCH, Chang GC, Han M, Komarnitsky P, Payumo F, Garrus JE, Close S, Park K. A Randomized Phase 2 Study Comparing the Combination of Ficlatuzumab and Gefitinib with Gefitinib Alone in Asian Patients with Advanced Stage Pulmonary Adenocarcinoma. J Thorac Oncol 2016; 11:1736-44. [PMID: 27448761 DOI: 10.1016/j.jtho.2016.05.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/24/2016] [Accepted: 05/30/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION A randomized phase 2 study was designed to compare the combination of ficlatuzumab (AV-299), a humanized hepatocyte growth factor-neutralizing monoclonal antibody, plus gefitinib versus gefitinib monotherapy in a pulmonary adenocarcinoma population clinically enriched for EFGR tyrosine kinase inhibitor-sensitizing mutations. METHODS A total of 188 patients were randomized 1:1 to receive either gefitinib or ficlatuzumab plus gefitinib treatment. Patients who demonstrated disease control in the single-agent gefitinib arm were allowed to cross over to ficlatuzumab plus gefitinib treatment upon disease progression. Molecular analyses included tumor EGFR mutation status and retrospective proteomic testing using VeriStrat, a multivariate test based on mass spectrometry. RESULTS The addition of ficlatuzumab to gefitinib did not provide significant improvement over gefitinib monotherapy for the primary end point of overall response rate or the secondary end points of progression-free survival and overall survival. In the subgroup classified as VeriStrat poor, the addition of ficlatuzumab to gefitinib showed significant improvement in both progression-free survival and overall survival in both the intent-to-treat population and the subgroup with EGFR tyrosine kinase inhibitor-sensitizing mutations. For all patients, the most frequent adverse events were diarrhea, dermatitis acneiform, and paronychia. CONCLUSIONS Although the trial showed no significant benefit from the addition of ficlatuzumab to gefitinib in the overall population of Asian patients with advanced-stage pulmonary adenocarcinoma, the biomarker data suggest that patients classified as VeriStrat poor may benefit from ficlatuzumab combination therapy.
Collapse
Affiliation(s)
- Tony Shu Kam Mok
- State Key Laboratory of South China, Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, Republic of China.
| | | | - Wu-Chou Su
- National Cheng Kung University Hospital, Tainan City, Republic of China
| | | | - James Chi-Hsin Yang
- National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China
| | - Gee-Chen Chang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Republic of China
| | - May Han
- AVEO Oncology, Cambridge, Massachusetts
| | | | | | | | | | - Keunchil Park
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
238
|
Ugolini A, Kenigsberg M, Rak A, Vallée F, Houtmann J, Lowinski M, Capdevila C, Khider J, Albert E, Martinet N, Nemecek C, Grapinet S, Bacqué E, Roesner M, Delaisi C, Calvet L, Bonche F, Semiond D, Egile C, Goulaouic H, Schio L. Discovery and Pharmacokinetic and Pharmacological Properties of the Potent and Selective MET Kinase Inhibitor 1-{6-[6-(4-Fluorophenyl)-[1,2,4]triazolo[4,3-b]pyridazin-3-ylsulfanyl]benzothiazol-2-yl}-3-(2-morpholin-4-ylethyl)urea (SAR125844). J Med Chem 2016; 59:7066-74. [DOI: 10.1021/acs.jmedchem.6b00280] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Manfred Roesner
- Sanofi-Aventis Germany GmbH, Industriepark
Hoechst, 65926 Frankfurt
am Main, Germany
| | | | | | | | - Dorothée Semiond
- Disposition,
Safety and Animal Research, Sanofi, 1 Avenue Pierre Brossolette, 91385 Chilly-Mazarin, France
| | | | | | | |
Collapse
|
239
|
Zheng D, Wang R, Ye T, Yu S, Hu H, Shen X, Li Y, Ji H, Sun Y, Chen H. MET exon 14 skipping defines a unique molecular class of non-small cell lung cancer. Oncotarget 2016; 7:41691-41702. [PMID: 27223439 PMCID: PMC5173088 DOI: 10.18632/oncotarget.9541] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/08/2016] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Recurrent MET exon 14 splicing has been revealed in lung cancers and is a promising therapeutic target. Because we have limited knowledge about the natural history of MET mutant tumors, the current study was aiming to determine the clinical and pathological characteristics in non-small cell lung cancers (NSCLC). RESULTS Twenty-three patients (1.3%) were positive for MET exon 14 skipping. Patients with MET exon 14 skipping displayed unique characteristics: female, non-smokers, earlier pathology stage and older age. MET exon 14 skipping indicated an early event as other drivers in lung cancer, while MET copy number gain was more likely a late event in lung cancer. Overall survival (OS) of patients harboring MET exon 14 skipping was longer than patients with KRAS mutation. Almost four-fifths of the lung tumors with MET exon 14 skipping had EGFR and/or HER2 gene copy number gains. EGFR inhibitor showed moderate antitumor activity in treatment of a patient harboring MET exon 14 skipping. PATIENTS AND METHODS From October 2007 to June 2013, we screened 1770 patients with NSCLC and correlated MET status with clinical pathologic characteristics and mutations in EGFR, KRAS, BRAF, HER2, and ALK. Quantitative Real-Time PCR was used to detect MET gene copy number gain. Immunohistochemistry (IHC) was also performed to screen MET exon 14 skipping. Clinicopathological characteristics and survival information were analyzed. CONCLUSIONS MET exon 14 skipping was detected in 1.3% (23/1770) of the Chinese patients with NSCLC. MET exon 14 skipping defined a new molecular subset of NSCLC with identifiable clinical characteristics. The therapeutic EGFR inhibitors might be an alternative treatment for patients with MET mutant NSCLC.
Collapse
Affiliation(s)
- Difan Zheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rui Wang
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Ye
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Su Yu
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Research Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haichuan Hu
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Xuxia Shen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuan Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hongbin Ji
- Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, China
| | - Yihua Sun
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
240
|
Zhang Y, Du Z, Zhang M. Biomarker development in MET-targeted therapy. Oncotarget 2016; 7:37370-37389. [PMID: 27013592 PMCID: PMC5095083 DOI: 10.18632/oncotarget.8276] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 03/14/2016] [Indexed: 12/16/2022] Open
Abstract
Activation of the MET receptor tyrosine kinase by its ligand, hepatocyte growth factor (HGF), has been implicated in a variety of cellular processes, including cell proliferation, survival, migration, motility and invasion, all of which may be enhanced in human cancers. Aberrantly activated MET/HGF signaling correlates with tumorigenesis and metastasis, and is regarded as a robust target for the development of novel anti-cancer treatments. Various clinical trials were conducted to evaluate the safety and efficacy of selective HGF/MET inhibitors in cancer patients. There is currently no optimal or standardized method for accurate and reliable assessment of MET levels, or other biomarkers that are predictive of the patient response to MET-targeted therapeutics. In this review, we discuss the importance of accurate HGF/MET signal detection as a predictive biomarker to guide patient selection for clinical trials of MET-targeted therapies in human cancers.
Collapse
Affiliation(s)
- Yanni Zhang
- Amgen Biopharmaceutical Research and Development (Shanghai) Co., Ltd, Shanghai, China
| | - Zhiqiang Du
- Amgen Biopharmaceutical Research and Development (Shanghai) Co., Ltd, Shanghai, China
| | - Mingqiang Zhang
- Amgen Biopharmaceutical Research and Development (Shanghai) Co., Ltd, Shanghai, China
| |
Collapse
|
241
|
Efficacy and Safety Results From a Phase II, Placebo-Controlled Study of Onartuzumab Plus First-Line Platinum-Doublet Chemotherapy for Advanced Squamous Cell Non-Small-Cell Lung Cancer. Clin Lung Cancer 2016; 18:43-49. [PMID: 27461773 DOI: 10.1016/j.cllc.2016.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 05/26/2016] [Indexed: 11/23/2022]
Abstract
INTRODUCTION The treatment options for squamous cell non-small-cell lung cancer (NSCLC) are limited. We assessed the efficacy and safety of onartuzumab plus platinum-doublet chemotherapy in previously untreated advanced squamous cell NSCLC. PATIENTS AND METHODS The patients were randomized to receive onartuzumab plus paclitaxel plus carboplatin/cisplatin (n = 55) or placebo plus paclitaxel plus carboplatin/cisplatin (n = 54). Randomization was stratified by MET diagnostic status: MET immunohistochemistry (IHC)-positive (MET IHC 3+/2+) or MET IHC-negative (MET IHC 1+/0). The co-primary endpoints were investigator-assessed progression-free survival in the intent-to-treat and the MET IHC+ populations. RESULTS The risk of disease progression or death was similar between the 2 treatment arms in both the intent-to-treat (stratified hazard ratio, 0.95; 95% confidence interval, 0.63-1.43) and MET IHC+ populations (unstratified hazard ratio, 1.27; 95% confidence interval, 0.69-2.32). Comparable results were obtained for overall survival and the objective response rate. In all safety-evaluable patients, the grade 3 to 5 adverse events occurring at a > 5% greater incidence in the onartuzumab-containing versus the placebo-containing arm were neutropenia (14.8% vs. 5.8%) and pulmonary embolism (5.6% vs. 0%). Eight patients died as a result of adverse events: 1 case each of pneumonitis, pneumonia, cardiac failure, and unexplained death in the onartuzumab arm and 1 case each of hemorrhage, cardiac arrest, hemoptysis, and febrile neutropenia in the placebo arm. CONCLUSION Studies using alternative assays of MET activation might help to clarify the role of onartuzumab. However, with the lack of clinical activity seen in the present study, the development of onartuzumab for squamous cell NSCLC will not be pursued further.
Collapse
|
242
|
Noonan SA, Berry L, Lu X, Gao D, Barón AE, Chesnut P, Sheren J, Aisner DL, Merrick D, Doebele RC, Varella-Garcia M, Camidge DR. Identifying the Appropriate FISH Criteria for Defining MET Copy Number-Driven Lung Adenocarcinoma through Oncogene Overlap Analysis. J Thorac Oncol 2016; 11:1293-1304. [PMID: 27262212 DOI: 10.1016/j.jtho.2016.04.033] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/07/2016] [Accepted: 04/27/2016] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Mesenchymal-epithelial transition factor gene (MET) gene copy number gain may be a predictive biomarker for mesenchymal-epithelial transition factor (MET) inhibition in lung cancer, but the most appropriate method and criteria for defining MET positivity are uncertain. METHODS MET copy number was assessed by fluorescence in situ hybridization in lung adenocarcinoma. Positivity criteria included mean MET per cell values greater than 5 (low [≥5 to <6], intermediate [≥6 to <7], and high [≥7]) and mean MET-to-chromosome 7 centromere ratios (MET/CEP7) of at least 1.8 (low [≥1.8 to ≤2.2], intermediate [>2.2 to <5], and high [≥5]). Associated clinical and molecular characteristics were captured. RESULTS Of 686 cases, 99 (14%) had a mean MET per cell value of 5 or greater, 52 of 1164 (4.5%) had a MET/CEP7 ratio of 1.8 or higher. Other oncogenic drivers (in EGFR, KRAS, anaplastic lymphoma receptor tyrosine kinase gene [ALK], erb-b2 receptor tyrosine kinase 2 gene [ERBB2], BRAF, NRAS, ROS1, or ret proto-oncogene [RET]) were detectable in 56% of the group with a mean MET per cell value of 5 or higher and 47% of the group with a MET/CEP7 ratio of 1.8 or higher, suggesting that many MET-positive cases are not truly MET addicted. The rates of concomitant drivers in the groups of patients in the low, indeterminate, and high categories of mean MET per cell were 32 of 52 (62%), 12 of 19 (63%), and 11 of 27 (41%) (p = 0.2), and the rates of concomitant drivers in the low, intermediate, and high categories of MET/CEP7 ratios were 15 of 29 (52%), 9 of 18 (50%), and 0 of 4 (0%), respectively (p = 0.04). A MET/CEP7 ratio of 1.8 or higher in the absence of other oncogenes was associated with a higher rate of adrenal metastases (p = 0.03) but not with never-smoking status. CONCLUSIONS A fluorescence in situ hybridization MET/CEP7 ratio of 5 or higher defined a "MET-positive" group with no oncogenic overlap. As this method and criteria are also associated with the highest response rate to MET inhibition, it represents the clearest definition of a MET copy number gain-addicted state. However, a MET-associated phenotype may also exist across cases with a MET/CEP7 of 1.8 or higher when no other oncogene overlap occurs.
Collapse
Affiliation(s)
- Sinéad A Noonan
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Lynne Berry
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Xian Lu
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Dexiang Gao
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Anna E Barón
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Patrick Chesnut
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Jamie Sheren
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Dara L Aisner
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Dan Merrick
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | - Robert C Doebele
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado
| | | | - D Ross Camidge
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, Colorado.
| |
Collapse
|
243
|
Salgia R. Mutation testing for directing upfront targeted therapy and post-progression combination therapy strategies in lung adenocarcinoma. Expert Rev Mol Diagn 2016; 16:737-49. [PMID: 27139190 PMCID: PMC4926789 DOI: 10.1080/14737159.2016.1181545] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Advances in the biology of non-small-cell lung cancer, especially adenocarcinoma, reveal multiple molecular subtypes driving oncogenesis. Accordingly, individualized targeted therapeutics are based on mutational diagnostics. Areas covered: Advances in strategies and techniques for individualized treatment, particularly of adenocarcinoma, are described through literature review. Approved therapies are established for some molecular subsets, with new driver mutations emerging that represent increasing proportions of patients. Actionable mutations are denovo oncogenic drivers or acquired resistance mediators, and mutational profiling is important for directing therapy. Patients should be monitored for emerging actionable resistance mutations. Liquid biopsy and associated multiplex diagnostics will be important means to monitor patients during treatment. Expert commentary: Outcomes with targeted agents may be improved by integrating mutation screens during treatment to optimize subsequent therapy. In order for this to be translated into impactful patient benefit, appropriate platforms and strategies need to be optimized and then implemented universally.
Collapse
Affiliation(s)
- Ravi Salgia
- a Department of Medical Oncology and Therapeutics Research , City of Hope , Duarte , CA , USA
| |
Collapse
|
244
|
Pyo JS, Kang G, Cho WJ, Choi SB. Clinicopathological significance and concordance analysis of c-MET immunohistochemistry in non-small cell lung cancers: A meta-analysis. Pathol Res Pract 2016; 212:710-6. [PMID: 27465837 DOI: 10.1016/j.prp.2016.05.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the clinicopathological significance and concordance rate of c-MET immunohistochemistry (IHC) in non-small cell lung cancer (NSCLC) through meta-analysis and diagnostic test accuracy review. METHODS The current study included 4454 NSCLC cases of 22 eligible studies. The meta-analysis examined the correlation between c-MET IHC expression and clinicopathological parameters. We investigated concordance rate between c-MET IHC and genetic alteration and performed subgroup analysis based on c-MET IHC cut-off value. RESULTS The estimated positive rate of c-MET IHC was 0.440 (95% confidence interval [CI] 0.355-0.529). The positive rate of c-MET IHC was significantly high in non-squamous cell carcinomas and tumors with stage III-IV. However, there was no significant difference between c-MET IHC positivity and sex, smoking, and lymph node metastasis. The c-MET IHC positivity was significantly correlated with poor overall survival (hazard ratio 1.551, 95% CI 1.101-2.184). In c-MET IHC-positive and negative groups, the concordance rate was 0.941 (95% CI 0.885-0.971) and 0.300 (95% CI 0.196-0.429), respectively. The pooled sensitivity and specificity of the high cut-off subgroup for c-MET IHC was 1.00 (95% CI 0.92-1.00) and 0.78 (95% CI 0.75-0.81), respectively. The diagnostic odds ratio and the area under curve on summary receiver operating characteristic curve were 76.56 (95% CI 8.23-712.41) and 0.9949, respectively. CONCLUSION The c-MET IHC could be useful for screening of c-MET genetic alteration in NSCLC patients. Detailed criteria for c-MET IHC evaluation are necessary to determine how to best apply this approach in daily practice.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Guhyun Kang
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Won Jin Cho
- Department of Urology, Chosun University Hospital, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Sang Bong Choi
- Department of Internal Medicine, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea.
| |
Collapse
|
245
|
Casadevall D, Gimeno J, Clavé S, Taus Á, Pijuan L, Arumí M, Lorenzo M, Menéndez S, Cañadas I, Albanell J, Serrano S, Espinet B, Salido M, Arriola E. MET expression and copy number heterogeneity in nonsquamous non-small cell lung cancer (nsNSCLC). Oncotarget 2016; 6:16215-26. [PMID: 26041880 PMCID: PMC4599265 DOI: 10.18632/oncotarget.3976] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/05/2015] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE We aimed to assess MET intratumoral heterogeneity and its potential impact on biomarker-based patient selection as well as potential surrogate biomarkers of MET activation. METHODS Our study included 120 patients with non-squamous Non-small-cell Lung Cancer (nsNSCLC), of which 47 were incorporated in tissue microarrays (TMA). Four morphologically distinct tumor areas were selected to assess MET heterogeneity. MET positivity by immunohistochemistry (IHC) was defined as an above-median H-score and by +2/+3 staining intensity in >50% of tumor cells (Metmab criteria). MET FISH positivity was defined by MET/CEP7 ratio ≥ 2.0 and/or MET ≥ 5.0. MET staining pattern (cytoplasmic vs. membranous) and mesenchymal markers were investigated as surrogates of MET activation. RESULTS Median MET H-score was 140 (range 0-400) and 47.8% of patients were MET positive by Metmab criteria. Eight cases (6.8%) were MET FISH positive and showed higher H-scores (p = 0.021). MET positivity by IHC changed in up to 40% of cases among different tumor areas, and MET amplification in 25-50%. Cytoplasmic MET staining and positivity for vimentin predicted poor survival (p = 0.042 and 0.047, respectively). CONCLUSIONS MET status is highly heterogeneous among different nsNSCLC tumor areas, hindering adequate patient selection for MET-targeted therapies. MET cytoplasmic staining and vimentin might represent surrogate markers for MET activation.
Collapse
Affiliation(s)
- David Casadevall
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Javier Gimeno
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Sergi Clavé
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain.,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Álvaro Taus
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - Lara Pijuan
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Miriam Arumí
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - Marta Lorenzo
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Silvia Menéndez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Israel Cañadas
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Joan Albanell
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain.,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Blanca Espinet
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain.,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marta Salido
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain.,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Edurne Arriola
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain.,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
246
|
Scientific Advances in Lung Cancer 2015. J Thorac Oncol 2016; 11:613-638. [DOI: 10.1016/j.jtho.2016.03.012] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023]
|
247
|
Li Y, Li W, He Q, Xu Y, Ren X, Tang X, Wen X, Yang X, Sun Y, Zeng J, Yun J, Liu N, Ma J. Prognostic value of MET protein overexpression and gene amplification in locoregionally advanced nasopharyngeal carcinoma. Oncotarget 2016; 6:13309-19. [PMID: 25965822 PMCID: PMC4537016 DOI: 10.18632/oncotarget.3751] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/08/2015] [Indexed: 11/25/2022] Open
Abstract
This study assessed the incidence and prognostic value of MET protein overexpression and gene amplification in locoregionally advanced nasopharyngeal carcinoma (NPC). Specimens from 376 consecutive patients with locoregionally advanced NPC were subjected to immunohistochemistry to analyze MET protein expression and fluorescence in situ hybridization to assess MET amplification status. In total, 139/376 (37.0%) patients had MET protein overexpression; of whom, 7/139 (5.0%) had MET amplification. MET overexpression was significantly associated with locoregional failure (P = 0.009), distant metastasis (P = 0.006) and death (P < 0.001); MET amplification was significantly associated with death (P = 0.021). A positive correlation was observed between MET copy number status and MET protein expression (r = 0.629, P < 0.001). Multivariate analysis demonstrated MET overexpression was an independent prognostic factor for overall survival (OS; HR, 1.99; 95% CI, 1.38-2.87; P < 0.001) and disease-free survival (DFS; HR, 1.85; 95% CI, 1.33-2.57; P < 0.001), and MET amplification was independently associated with poorer OS (HR, 4.24; 95% CI, 1.78-10.08; P < 0.001) and DFS (HR, 5.44; 95% CI, 2.44-12.09; P < 0.001). In conclusion, MET protein overexpression and gene amplification are independent prognostic factors for OS and DFS in locoregionally advanced nasopharyngeal carcinoma, and may provide therapeutic biomarkers to identify patients in whom MET inhibitors may be beneficial.
Collapse
Affiliation(s)
- Yingqin Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenfei Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Qingmei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yafei Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xianyue Ren
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xinran Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xin Wen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xiaojing Yang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ying Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jing Zeng
- Department of Pathology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingping Yun
- Department of Pathology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Na Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jun Ma
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| |
Collapse
|
248
|
Tang Q, Wang L, Tu Y, Zhu W, Luo R, Tu Q, Wang P, Wu C, Gong P, Zheng P. Discovery of novel pyrrolo[2,3-b]pyridine derivatives bearing 1,2,3-triazole moiety as c-Met kinase inhibitors. Bioorg Med Chem Lett 2016; 26:1680-4. [DOI: 10.1016/j.bmcl.2016.02.059] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 01/30/2016] [Accepted: 02/19/2016] [Indexed: 10/22/2022]
|
249
|
Sholl LM, Aisner DL, Varella-Garcia M, Berry LD, Dias-Santagata D, Wistuba II, Chen H, Fujimoto J, Kugler K, Franklin WA, Iafrate AJ, Ladanyi M, Kris MG, Johnson BE, Bunn PA, Minna JD, Kwiatkowski DJ. Multi-institutional Oncogenic Driver Mutation Analysis in Lung Adenocarcinoma: The Lung Cancer Mutation Consortium Experience. J Thorac Oncol 2016; 10:768-777. [PMID: 25738220 DOI: 10.1097/jto.0000000000000516] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Molecular genetic analyses of lung adenocarcinoma have recently become standard of care for treatment selection. The Lung Cancer Mutation Consortium was formed to enable collaborative multi-institutional analyses of 10 potential oncogenic driver mutations. Technical aspects of testing and clinicopathologic correlations are presented. METHODS Mutation testing in at least one of the eight genes (epidermal growth factor receptor [EGFR], KRAS, ERBB2, AKT1, BRAF, MEK1, NRAS, and PIK3CA) using SNaPshot, mass spectrometry, Sanger sequencing+/- peptide nucleic acid and/or sizing assays, along with anaplastic lymphoma kinase (ALK) and/or MET fluorescence in situ hybridization, were performed in six labs on 1007 patients from 14 institutions. RESULTS In all, 1007 specimens had mutation analysis performed, and 733 specimens had all 10 genes analyzed. Mutation identification rates did not vary by analytic method. Biopsy and cytology specimens were inadequate for testing in 26% and 35% of cases compared with 5% of surgical specimens. Among the 1007 cases with mutation analysis performed, EGFR, KRAS, ALK, and ERBB2 alterations were detected in 22%, 25%, 8.5%, and 2.4% of cases, respectively. EGFR mutations were highly associated with female sex, Asian race, and never-smoking status; and less strongly associated with stage IV disease, presence of bone metastases, and absence of adrenal metastases. ALK rearrangements were strongly associated with never-smoking status and more weakly associated with presence of liver metastases. ERBB2 mutations were strongly associated with Asian race and never-smoking status. Two mutations were seen in 2.7% of samples, all but one of which involved one or more of PIK3CA, ALK, or MET. CONCLUSION Multi-institutional molecular analysis across multiple platforms, sample types, and institutions can yield consistent results and novel clinicopathological observations.
Collapse
Affiliation(s)
- Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Dara L Aisner
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Marileila Varella-Garcia
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Lynne D Berry
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Heidi Chen
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Kelly Kugler
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark G Kris
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bruce E Johnson
- Department of Medicine, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Paul A Bunn
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; Lung Cancer Mutation Consortium, University of Colorado Cancer Center, Aurora, Colorado
| | | | - David J Kwiatkowski
- Department of Medicine, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | | |
Collapse
|
250
|
Ariyawutyakorn W, Saichaemchan S, Varella-Garcia M. Understanding and Targeting MET Signaling in Solid Tumors - Are We There Yet? J Cancer 2016; 7:633-49. [PMID: 27076844 PMCID: PMC4829549 DOI: 10.7150/jca.12663] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 01/22/2016] [Indexed: 12/22/2022] Open
Abstract
The MET signaling pathway plays an important role in normal physiology and its deregulation has proved critical for development of numerous solid tumors. Different technologies have been used to investigate the genomic and proteomic status of MET in cancer patients and its association with disease prognosis. Moreover, with the development of targeted therapeutic drugs, there is an urgent need to identify potential biomarkers for selection of patients who are more likely to derive benefit from these agents. Unfortunately, the variety of technical platforms and analysis criteria for diagnosis has brought confusion to the field and a lack of agreement in the evaluation of MET status as a prognostic or predictive marker for targeted therapy agents. We review the molecular mechanisms involved in the deregulation of the MET signaling pathway in solid tumors, the different technologies used for diagnosis, and the main factors that affect the outcome, emphasizing the urge for completing analytical and clinical validation of these tests. We also review the current clinical studies with MET targeted agents, which mostly focus on lung cancer.
Collapse
Affiliation(s)
- Witthawat Ariyawutyakorn
- 1. Faculty of Medicine, Chiang Mai University, 110 Intavarorod Rd., Muang, Chiang Mai, Thailand 50200
- 3. Department of Medicine, University of Colorado, Anschutz Medical Campus, 12801 East 17th Ave, RC1 South, L18-8118, Mail Stop 8117, Aurora, Colorado, USA 80045
| | - Siriwimon Saichaemchan
- 2. Division of Oncology, Department of Medicine, Phramongkutklao Hospital and College of Medicine, 315 Phayathai Rd., Ratchathewi, Bangkok, Thailand 10400
- 3. Department of Medicine, University of Colorado, Anschutz Medical Campus, 12801 East 17th Ave, RC1 South, L18-8118, Mail Stop 8117, Aurora, Colorado, USA 80045
| | - Marileila Varella-Garcia
- 3. Department of Medicine, University of Colorado, Anschutz Medical Campus, 12801 East 17th Ave, RC1 South, L18-8118, Mail Stop 8117, Aurora, Colorado, USA 80045
| |
Collapse
|