151
|
Jurmeister P, Schöler A, Arnold A, Klauschen F, Lenze D, Hummel M, Schweizer L, Bläker H, Pfitzner BM, Mamlouk S, Sers C, Denkert C, Stichel D, Frost N, Horst D, von Laffert M, Capper D. DNA methylation profiling reliably distinguishes pulmonary enteric adenocarcinoma from metastatic colorectal cancer. Mod Pathol 2019; 32:855-865. [PMID: 30723296 DOI: 10.1038/s41379-019-0207-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/27/2018] [Accepted: 12/29/2018] [Indexed: 12/20/2022]
Abstract
Pulmonary enteric adenocarcinoma is a rare non-small cell lung cancer subtype. It is poorly characterized and cannot be distinguished from metastatic colorectal or upper gastrointestinal adenocarcinomas by means of routine pathological methods. As DNA methylation patterns are known to be highly tissue specific, we aimed to develop a methylation-based algorithm to differentiate these entities. To this end, genome-wide methylation profiles of 600 primary pulmonary, colorectal, and upper gastrointestinal adenocarcinomas obtained from The Cancer Genome Atlas and the Gene Expression Omnibus database were used as a reference cohort to train a machine learning algorithm. The resulting classifier correctly classified all samples from a validation cohort consisting of 680 primary pulmonary, colorectal and upper gastrointestinal adenocarcinomas, demonstrating the ability of the algorithm to reliably distinguish these three entities. We then analyzed methylation data of 15 pulmonary enteric adenocarcinomas as well as four pulmonary metastases and four primary colorectal adenocarcinomas with the algorithm. All 15 pulmonary enteric adenocarcinomas were reliably classified as primary pulmonary tumors and all four metastases as well as all four primary colorectal cancer samples were identified as colorectal adenocarcinomas. In a t-distributed stochastic neighbor embedding analysis, the pulmonary enteric adenocarcinoma samples did not form a separate methylation subclass but rather diffusely intermixed with other pulmonary cancers. Additional characterization of the pulmonary enteric adenocarcinoma series using fluorescence in situ hybridization, next-generation sequencing and copy number analysis revealed KRAS mutations in nine of 15 samples (60%) and a high number of structural chromosomal changes. Except for an unusually high rate of chromosome 20 gain (67%), the molecular data was mostly reminiscent of standard pulmonary adenocarcinomas. In conclusion, we provide sound evidence of the pulmonary origin of pulmonary enteric adenocarcinomas and in addition provide a publicly available machine learning-based algorithm to reliably distinguish these tumors from metastatic colorectal cancer.
Collapse
Affiliation(s)
- Philipp Jurmeister
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany. .,Charité Comprehensive Cancer Center (CCCC), Berlin, Germany.
| | - Anne Schöler
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Alexander Arnold
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Frederick Klauschen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dido Lenze
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Michael Hummel
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Leonille Schweizer
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hendrik Bläker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Berit Maria Pfitzner
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Soulafa Mamlouk
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Sers
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carsten Denkert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Damian Stichel
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nikolaj Frost
- Department of Infectious Diseases and Pneumonology, Charité University Hospital Berlin, Berlin, Germany
| | - David Horst
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maximilian von Laffert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - David Capper
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
152
|
Yu J, Wang Q, Xue P, Zheng L, Mo J, Chen L, Yin M, Huang Y, Bao Y, Ding F. A model for the impact of FFPE section thickness on gene copy number measurement by FISH. Sci Rep 2019; 9:7518. [PMID: 31101839 PMCID: PMC6525178 DOI: 10.1038/s41598-019-44015-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 05/07/2019] [Indexed: 01/16/2023] Open
Abstract
Fluorescent in situ hybridization (FISH) assays to detect gene amplification such as HER2 or MET in tumors are used for prognosis evaluation and selection of targeted therapies. Although FISH guidelines recommended 4~6 μm FFPE sections, many laboratories use 2~3 μm sections, which is a common practice for H&E staining and immunohistochemistry. A former study concluded that section thickness did not affect FISH results. We found, however, that thinner FFPE sections may lead to false negative results for gene amplification. A mathematic model was constructed and cell-line based controls with known gene copy number were prepared, and the model had a reasonable fit with the experimental data. The model revealed that even when counting the apparently full-sized nuclear images, many of them have partial volumes, which leads to under-estimation of gene copy number. Therefore, improperly thinner sections are prone to give false negative results, and thicker sections give a better approximation to the true value. The discrepancy between this and the former study was discussed. In summary, the model applies generally to FISH/ISH detection of gene copy number, and section thickness is an important parameter to control for precision medicine research, assay development, clinical trials and daily practice in pathology laboratory.
Collapse
Affiliation(s)
- Jiyan Yu
- Department of Biotechnology and Biomedicine, and Zhejiang Provincial Key Laboratory of Applied Enzymology, Jiaxing ACCB Diagnostics, Yangze Delta Regional Institute of Tsinghua University, Zhejiang, Jiaxing, 314006, China
| | - Qi Wang
- Department of Biotechnology and Biomedicine, and Zhejiang Provincial Key Laboratory of Applied Enzymology, Jiaxing ACCB Diagnostics, Yangze Delta Regional Institute of Tsinghua University, Zhejiang, Jiaxing, 314006, China
| | - Pu Xue
- Department of Biotechnology and Biomedicine, and Zhejiang Provincial Key Laboratory of Applied Enzymology, Jiaxing ACCB Diagnostics, Yangze Delta Regional Institute of Tsinghua University, Zhejiang, Jiaxing, 314006, China
| | - Li Zheng
- Department of Research Center, The Second Hospital of Jiaxing, Jiaxing, China
| | - Juanfen Mo
- Department of Research Center, The Second Hospital of Jiaxing, Jiaxing, China
| | - Liangye Chen
- Department of Pathology, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, and Medical College of Jiaxing University, Jiaxing, 314000, China
| | - Manxiang Yin
- Department of Pathology, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, and Medical College of Jiaxing University, Jiaxing, 314000, China
| | - Yueyan Huang
- Department of Pathology, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, and Medical College of Jiaxing University, Jiaxing, 314000, China
| | - Yi Bao
- Department of Research Center, The Second Hospital of Jiaxing, Jiaxing, China.
| | - Feng Ding
- Department of Biotechnology and Biomedicine, and Zhejiang Provincial Key Laboratory of Applied Enzymology, Jiaxing ACCB Diagnostics, Yangze Delta Regional Institute of Tsinghua University, Zhejiang, Jiaxing, 314006, China.
| |
Collapse
|
153
|
Singh R, Peng S, Viswanath P, Sambandam V, Shen L, Rao X, Fang B, Wang J, Johnson FM. Non-canonical cMet regulation by vimentin mediates Plk1 inhibitor-induced apoptosis. EMBO Mol Med 2019; 11:e9960. [PMID: 31040125 PMCID: PMC6505578 DOI: 10.15252/emmm.201809960] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/28/2019] [Accepted: 03/12/2019] [Indexed: 12/26/2022] Open
Abstract
To address the need for improved systemic therapy for non-small-cell lung cancer (NSCLC), we previously demonstrated that mesenchymal NSCLC was sensitive to polo-like kinase (Plk1) inhibitors, but the mechanisms of resistance in epithelial NSCLC remain unknown. Here, we show that cMet was differentially regulated in isogenic pairs of epithelial and mesenchymal cell lines. Plk1 inhibition inhibits cMet phosphorylation only in mesenchymal cells. Constitutively active cMet abrogates Plk1 inhibitor-induced apoptosis. Likewise, cMet silencing or inhibition enhances Plk1 inhibitor-induced apoptosis. Cells with acquired resistance to Plk1 inhibitors are more epithelial than their parental cells and maintain cMet activation after Plk1 inhibition. In four animal NSCLC models, mesenchymal tumors were more sensitive to Plk1 inhibition alone than were epithelial tumors. The combination of cMet and Plk1 inhibition led to regression of tumors that did not regrow when drug treatment was stopped. Plk1 inhibition did not affect HGF levels but did decrease vimentin phosphorylation, which regulates cMet phosphorylation via β1-integrin. This research defines a heretofore unknown mechanism of ligand-independent activation of cMet downstream of Plk1 and an effective combination therapy.
Collapse
Affiliation(s)
- Ratnakar Singh
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shaohua Peng
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pavitra Viswanath
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Vaishnavi Sambandam
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiayu Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faye M Johnson
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
154
|
Ando R, Fujino M, Kominami-Kiriyama A, Ito A, Koide T, Ito M. Mesenchymal–epithelial transition gene amplification and protein overexpression in stage IV pulmonary adenocarcinoma. Jpn J Clin Oncol 2019; 49:755-761. [DOI: 10.1093/jjco/hyz060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/17/2019] [Accepted: 04/07/2019] [Indexed: 12/27/2022] Open
Abstract
Abstract
Background
In non-small cell lung cancer (NSCLC), MET gene copy number gain, including gene amplification and chromosome 7 polysomy, is reportedly associated with patient prognosis. Although relationship between MET copy number gain and poor prognosis has been suggested in surgically resected non-small cell lung cancer, the clinical significance of MET copy number gain and protein overexpression in patients with advanced unresectable tumor is unclear.
Methods
We assessed MET copy number gain and protein expression using fluorescence in situ hybridization and immunohistochemistry in 88 patients with clinical stage IV pulmonary adenocarcinoma receiving chemotherapy, immunotherapy or palliative care.
Results
We found MET amplification, polysomy 7 and high MET protein expression in 10.2, 18.2 and 62.5% of 88 cases, respectively. Gene amplification and high protein expression were not significantly associated. A univariate analysis showed that MET amplification-positive patients had increased overall survival (HR 0.335, 95% CI: 0.119–0.945; P = 0.0388). Although it was not statistically significant in the multivariate analysis of the whole cohort, with the removal of patients who did not receive any treatment other than palliative care, MET amplification independently improved the overall survival (HR 0.178, 95% CI: 0.041–0.770; P = 0.0209). Chromosome 7 polysomy and high MET protein expression did not affect the overall survival.
Conclusions
Although MET amplification-positive tumor is considered aggressive, our results suggest that it has a more favorable prognosis than amplification-negative cases in stage IV pulmonary adenocarcinoma with medical treatment.
Collapse
Affiliation(s)
- Ryota Ando
- Department of Pathology, Japanese Red Cross Nagoya First Hospital, 3-35, Nagoya 4538511, Japan
| | - Masahiko Fujino
- Department of Cytology and Molecular Pathology, Japanese Red Cross Nagoya First Hospital, 3-35, Nagoya 4538511, Japan
| | - Ayami Kominami-Kiriyama
- Department of Pathology, Japanese Red Cross Nagoya First Hospital, 3-35, Nagoya 4538511, Japan
| | - Ai Ito
- Department of Pathology, Japanese Red Cross Nagoya First Hospital, 3-35, Nagoya 4538511, Japan
| | - Tomomi Koide
- Department of Pathology, Japanese Red Cross Nagoya First Hospital, 3-35, Nagoya 4538511, Japan
| | - Masafumi Ito
- Department of Pathology, Japanese Red Cross Nagoya First Hospital, 3-35, Nagoya 4538511, Japan
| |
Collapse
|
155
|
Hong DS, LoRusso P, Hamid O, Janku F, Kittaneh M, Catenacci DVT, Chan E, Bekaii-Saab T, Gadgeel SM, Loberg RD, Amore BM, Hwang YC, Tang R, Ngarmchamnanrith G, Kwak EL. Phase I Study of AMG 337, a Highly Selective Small-molecule MET Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res 2019; 25:2403-2413. [PMID: 30425090 PMCID: PMC6892342 DOI: 10.1158/1078-0432.ccr-18-1341] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/28/2018] [Accepted: 11/08/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE This first-in-human, open-label phase I study evaluated AMG 337, an oral, highly selective small-molecule inhibitor of MET in advanced solid tumors.Patients and Methods: Patients enrolled into dose-escalation cohorts received AMG 337 up to 400 mg once daily or up to 250 mg twice daily, following a modified 3+3+3 design. Dose expansion was conducted in MET-amplified patients at the maximum tolerated dose (MTD). Primary endpoints included assessment of adverse events (AEs), establishment of the MTD, and pharmacokinetics; clinical response was a secondary endpoint. RESULTS The safety analysis set included 111 patients who received ≥1 dose of AMG 337. Thirteen patients had ≥1 AE qualifying as dose-limiting toxicity. The MTD was determined to be 300 mg once daily; the MTD for twice-daily dosing was not reached. Most frequent treatment-related AEs were headache (63%) and nausea (31%). Grade ≥3 treatment-related AEs occurred in 23 patients (21%), most commonly headache (n = 6) and fatigue (n = 5). Maximum plasma concentration occurred at 3.0 hours following 300-mg once-daily dosing, indicating AMG 337 absorption soon after treatment. Objective response rate was 9.9% (11/111; 95% CI, 5.1%-17.0%) in all patients and 29.6% (8/27; 95% CI, 13.8%-50.2%) in MET-amplified patients; median (range) duration of response was 202 (51-1,430+) days in all patients and 197 (64-1,430+) days in MET-amplified patients. CONCLUSIONS Oral AMG 337 was tolerated with manageable toxicities, with an MTD and recommended phase II dose of 300 mg once daily. The promising response rate observed in patients with heavily pretreated MET-amplified tumors warrants further investigation.See related commentary by Ma, p. 2375.
Collapse
Affiliation(s)
- David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Omid Hamid
- Melanoma Center, The Angeles Clinic and Research Institute, Los Angeles, California
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muaiad Kittaneh
- Hematology/Oncology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | | | - Emily Chan
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | | | - Shirish M Gadgeel
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
- Thoracic Oncology, Karmanos Cancer Institute, Detroit, Michigan
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan
| | | | - Benny M Amore
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., South San Francisco, California
| | - Yuying C Hwang
- Global Biostatistical Sciences, Amgen Inc., Thousand Oaks, California
| | - Rui Tang
- Global Biostatistical Sciences, Amgen Inc., Thousand Oaks, California
| | | | - Eunice L Kwak
- Hematology/Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| |
Collapse
|
156
|
Lai GGY, Lim TH, Lim J, Liew PJR, Kwang XL, Nahar R, Aung ZW, Takano A, Lee YY, Lau DPX, Tan GS, Tan SH, Tan WL, Ang MK, Toh CK, Tan BS, Devanand A, Too CW, Gogna A, Ong BH, Koh TPT, Kanesvaran R, Ng QS, Jain A, Rajasekaran T, Yuan J, Lim TKH, Lim AST, Hillmer AM, Lim WT, Iyer NG, Tam WL, Zhai W, Tan EH, Tan DSW. Clonal MET Amplification as a Determinant of Tyrosine Kinase Inhibitor Resistance in Epidermal Growth Factor Receptor-Mutant Non-Small-Cell Lung Cancer. J Clin Oncol 2019; 37:876-884. [PMID: 30676858 DOI: 10.1200/jco.18.00177] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2018] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Mesenchymal epithelial transition factor ( MET) activation has been implicated as an oncogenic driver in epidermal growth factor receptor ( EGFR)-mutant non-small-cell lung cancer (NSCLC) and can mediate primary and secondary resistance to EGFR tyrosine kinase inhibitors (TKI). High copy number thresholds have been suggested to enrich for response to MET inhibitors. We examined the clinical relevance of MET copy number gain (CNG) in the setting of treatment-naive metastatic EGFR-mutant-positive NSCLC. PATIENTS AND METHODS MET fluorescence in situ hybridization was performed in 200 consecutive patients identified as metastatic treatment-naïve EGFR-mutant-positive. We defined MET-high as CNG greater than or equal to 5, with an additional criterion of MET/centromeric portion of chromosome 7 ratiο greater than or equal to 2 for amplification. Time-to-treatment failure (TTF) to EGFR TKI in patients identified as MET-high and -low was estimated by Kaplan-Meier method and compared using log-rank test. Multiregion single-nucleotide polymorphism array analysis was performed on 13 early-stage resected EGFR-mutant-positive NSCLC across 59 sectors to investigate intratumoral heterogeneity of MET CNG. RESULTS Fifty-two (26%) of 200 patients in the metastatic cohort were MET-high at diagnosis; 46 (23%) had polysomy and six (3%) had amplification. Median TTF was 12.2 months (95% CI, 5.7 to 22.6 months) versus 13.1 months (95% CI, 10.6 to 15.0 months) for MET-high and -low, respectively ( P = .566), with no significant difference in response rate regardless of copy number thresholds. Loss of MET was observed in three of six patients identified as MET-high who underwent postprogression biopsies, which is consistent with marked intratumoral heterogeneity in MET CNG observed in early-stage tumors. Suboptimal response (TTF, 1.0 to 6.4 months) to EGFR TKI was observed in patients with coexisting MET amplification (five [3.2%] of 154). CONCLUSION Although up to 26% of TKI-naïve EGFR-mutant-positive NSCLC harbor high MET CNG by fluorescence in situ hybridization, this did not significantly affect response to TKI, except in patients identified as MET-amplified. Our data underscore the limitations of adopting arbitrary copy number thresholds and the need for cross-assay validation to define therapeutically tractable MET pathway dysregulation in EGFR-mutant-positive NSCLC.
Collapse
Affiliation(s)
| | | | - John Lim
- 1 National Cancer Centre Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | - Mei-Kim Ang
- 1 National Cancer Centre Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | - Amit Jain
- 1 National Cancer Centre Singapore, Singapore
| | | | - Ju Yuan
- 3 Genome Institute of Singapore, Singapore
| | | | | | | | | | | | | | | | | | - Daniel S W Tan
- 1 National Cancer Centre Singapore, Singapore
- 3 Genome Institute of Singapore, Singapore
| |
Collapse
|
157
|
Rivalland G, Mitchell P, Murone C, Asadi K, Morey AL, Starmans M, Boutros PC, Walkiewicz M, Solomon B, Wright G, Knight S, John T. Mesenchyme to epithelial transition protein expression, gene copy number and clinical outcome in a large non-small cell lung cancer surgical cohort. Transl Lung Cancer Res 2019; 8:167-175. [PMID: 31106127 DOI: 10.21037/tlcr.2019.03.11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background In non-small cell lung cancer (NSCLC), mesenchyme to epithelial transition (MET) protein abundance increases with disease stage and is implicated in resistance to tyrosine kinase inhibitors. To better clarify the impact of MET overexpression on tumor behavior, we investigated a large cohort of patients who underwent curative surgical resection to determine whether MET gene amplification or protein abundance was prognostic. Methods Tissue microarrays (TMAs) were constructed using triplicate 1 mm cores of FFPE primary NSCLC specimens. TMAs underwent immunohistochemical (IHC) staining with the SP44 clone (Ventana) and cores were considered positive if >50% of tumor exhibited 2+ staining. The highest of triplicate values was used. MET gene amplification was detected using either SISH using Ventana's MET DNP probe or FISH using the D7S486/CEP 7 Abbott Probe. DNA was subjected to mutational profiling using Sequenom's LungCarta panel. Results Data from two institutions comprising 763 patients (516; 68%) male were generated, including 360 stage I, 226 stage II, 160 stage III and 18 resected stage IV. High MET protein expression was detected in 25% (193/763), and was significantly more common in adenocarcinomas than squamous cell carcinoma (P<0.01). MET gene copy number (GCN) correlated with high MET protein expression by IHC (P=0.01). Increased MET protein expression was associated with EGFR and KRAS mutations (P<0.01 for both). Once polysomy was excluded, true MET gene amplification was detected in only 8/763 (1%) of samples. In multivariate analysis, neither MET protein abundance nor GCN were correlated to overall patient survival. Conclusions MET expression by IHC and GCN amplification was not prognostic in this large Caucasian surgical series. MET's primary role remains as a therapeutic target.
Collapse
Affiliation(s)
- Gareth Rivalland
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Faculty of Medicine, University of Melbourne, Melbourne, Australia.,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada
| | - Paul Mitchell
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medical Oncology, Austin Health, Melbourne, Australia
| | - Carmel Murone
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Khashayer Asadi
- Department of Pathology, Austin Health, Melbourne, Australia
| | - Adrienne L Morey
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, Australia
| | - Maud Starmans
- Ontario Institute for Cancer Research, Toronto, Canada.,Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Paul C Boutros
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada.,Ontario Institute for Cancer Research, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Benjamin Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Gavin Wright
- Research and Education Lead in Lung Cancer, Victorian Comprehensive Cancer Centre, Parkville, Australia.,Department of Surgery, University of Melbourne, St Vincent's Hospital, Fitzroy, Australia
| | - Simon Knight
- Department of Thoracic Surgery, Austin Health, Melbourne, Australia
| | - Thomas John
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Faculty of Medicine, University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, Austin Health, Melbourne, Australia
| |
Collapse
|
158
|
Wang Y, Jiang Z, Xu C, Wang H, Tan L, Su J, Wang X, Jiang D, Hou Y, Song Q. Increased MET gene copy number negatively affects the survival of esophageal squamous cell carcinoma patients. BMC Cancer 2019; 19:240. [PMID: 30885149 PMCID: PMC6421677 DOI: 10.1186/s12885-019-5450-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 03/11/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUNDS Since Mesenchymal epithelial transition (MET) amplification has been regarded as a potential treatment target, the knowledge of its prevalence and prognostic importance is crucial. However, its clinical pathologic characteristics are not well known in esophageal squamous cell carcinoma (ESCC). METHODS We investigated MET gene status with fluorescence in situ hybridization (FISH) assay in 495 ESCC cases using tissue microarrays. Prognostic significance as well as correlations with various clinicopathological parameters was evaluated. RESULTS Among 495 patients, 28 (5.7%) cases were MET FISH positive, including 5 cases (1%) with true gene amplification. There were no statistically significant associations between MET FISH-positivity and clinicopathologic characteristics. A significantly poorer prognosis was observed in 28 patients with MET FISH-positivity (disease free survival/DFS, P < 0.001 and overall survival/OS, P = 0.001). Multivariate analysis revealed MET FISH-positivity was an independent prognostic factor for DFS (hazard ratio/HR, 1.953; 95% confidence interval/CI, 1.271-2.999; P = 0.002) and OS (HR, 1.926; 95% CI, 1.243-2.983; P = 0.003). MET FISH-positivity was associated with DFS (P = 0.022 and 0.020) and OS (P = 0.046 and 0.024) both in stage I-II ESCC and in stage III-IVa ESCC. No statistical significance (DFS, P = 0.492 and OS, P = 0.344) was detected between stage I-II ESCC with MET FISH-positivity and stage III-IVa ESCC with FISH-negativity. CONCLUSIONS Increased MET gene copy number is an independent prognostic factor in ESCC, and ESCC might have potentially been up-staged by increased MET gene copy number. The results indicate that increased MET gene copy number is a very promising parameter, in clinical therapy and follow-up plans.
Collapse
Affiliation(s)
- Yanqiu Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Zhengzeng Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hao Wang
- Department of Thoracic surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Lijie Tan
- Department of Thoracic surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xin Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Department of Pathology, School of Basic Medical Sciences & Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, People's Republic of China.
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
159
|
Rehman S, Dy GK. MET Inhibition in Non-Small Cell Lung Cancer. EUROPEAN MEDICAL JOURNAL 2019. [DOI: 10.33590/emj/10314675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cancer treatment paradigms have evolved over recent years with an emphasis on personalised medicine. Targeted agents are being used to improve treatment outcomes and quality of life. For the treatment of non-small cell lung cancer, several agents with unique genetic and epigenetic targets are available. To this extent, mesenchymal–epithelial transition (MET), a heterodimer receptor tyrosine kinase involved in embryogenesis and organogenesis, has been investigated as a potential target for biological agents. MET dysregulation can occur via different mechanisms and trigger tumourigenesis and disease spread. Besides driving the oncogenic dependence of cells, MET is also involved in acquired resistance to epidermal growth factor receptor inhibitors. As such, many small molecule kinase inhibitors and antibodies have been developed or are currently in different phases of clinical trials to counteract the MET-induced neoplastic activity. Some of these agents are selective while others are nonselective with multiple other potential targets. This article aims to present an overview of biological functioning of MET, its role in oncogenesis and resistance to treatment, and clinical studies evaluating MET inhibitors for treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Shabnam Rehman
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Grace K. Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
160
|
Reckamp KL, Frankel PH, Ruel N, Mack PC, Gitlitz BJ, Li T, Koczywas M, Gadgeel SM, Cristea MC, Belani CP, Newman EM, Gandara DR, Lara PN. Phase II Trial of Cabozantinib Plus Erlotinib in Patients With Advanced Epidermal Growth Factor Receptor ( EGFR)-Mutant Non-small Cell Lung Cancer With Progressive Disease on Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy: A California Cancer Consortium Phase II Trial (NCI 9303). Front Oncol 2019; 9:132. [PMID: 30915273 PMCID: PMC6421302 DOI: 10.3389/fonc.2019.00132] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/14/2019] [Indexed: 11/30/2022] Open
Abstract
Introduction: Mesenchymal epidermal transition and vascular endothelial growth factor pathways are important in mediating non-small cell lung cancer (NSCLC) tumorigenesis and epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) resistance. We hypothesized that treatment with cabozantinib plus erlotinib in EGFR mutation-positive NSCLC following progression on EGFR TKI therapy may allow tumors to overcome this resistance or restore sensitivity to therapy regardless of T790M status. Methods: Patients with advanced NSCLC, known EGFR mutation and progressive disease on an EGFR TKI immediately prior to enrollment without intervening therapy were enrolled. Patients received erlotinib 150 mg and cabozantinib 40 mg daily. The primary endpoint was evaluation of efficacy by objective response rate. Secondary endpoints included assessment of progression free survival (PFS), overall survival, change in tumor growth rate, safety and toxicity, and the evaluation of specific EGFR mutations and MET amplification in pre-treatment tissue and plasma. Results: Thirty-seven patients were enrolled at 4 centers. Four patients had partial response (10.8%) and 21 had stable disease (59.5%). A greater than 30% increase in tumor doubling time was observed in 79% of assessable patients (27/34). Median PFS was 3.6 months for all patients. Diarrhea (32%) was the most common grade 3 adverse event; 3 patients had asymptomatic grade 4 elevation of amylase and lipase. Conclusions: Combination erlotinib and cabozantinib demonstrates activity in a highly pretreated population of patients with EGFR mutation and progression on EGFR TKI. Further elucidation of beneficial patient subsets is warranted. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT01866410.
Collapse
Affiliation(s)
- Karen L. Reckamp
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Paul H. Frankel
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Nora Ruel
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Philip C. Mack
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, United States
| | | | - Tianhong Li
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, United States
| | | | - Shirish M. Gadgeel
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | | | | | - Edward M. Newman
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - David R. Gandara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, United States
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, United States
| |
Collapse
|
161
|
Esaki T, Hirai F, Makiyama A, Seto T, Bando H, Naito Y, Yoh K, Ishihara K, Kakizume T, Natsume K, Myers A, Doi T. Phase I dose-escalation study of capmatinib (INC280) in Japanese patients with advanced solid tumors. Cancer Sci 2019; 110:1340-1351. [PMID: 30724423 PMCID: PMC6447844 DOI: 10.1111/cas.13956] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
Capmatinib is a highly specific, potent and selective MET inhibitor. This was an open‐label, multicenter, dose‐escalation, phase I study conducted in Japanese patients with advanced solid tumors (not selected based on their MET status). The primary objective was to determine the maximum tolerated dose (MTD) and/or highest studied dose being safe. Secondary objectives included safety, pharmacokinetics and preliminary antitumor activity. Dose escalation was guided by a Bayesian Logistic Regression Model dependent on dose‐limiting toxicities (DLT) in cycle 1. Of 44 adult Japanese patients with confirmed advanced solid tumors enrolled, 29 received capmatinib capsules (doses ranging from 100 mg once daily [q.d.] to 600 mg twice daily [b.i.d.]) and 15 received tablets (200 mg b.i.d. and 400 mg b.i.d.). DLT occurred in two patients: grade 2 suicidal ideation (600 mg b.i.d. capsule) and grade 3 depression (400 mg b.i.d. tablet). MTD was not reached. The highest studied dose determined to be safe as tablet was 400 mg b.i.d., whereas it is not yet determined for capsules. Most common adverse events suspected to be drug‐related were increased blood creatinine, nausea, decreased appetite, vomiting and diarrhea. Following repeated daily dosing up to day 15 by q.d. or b.i.d. regimen using capsules, median time to reach maximum plasma drug concentration (Tmax) was 1.0‐4.0 hours; absorption was more rapid after dosing using tablets, with median Tmax of 1.0 hour on both days 1 and 15. Eight patients had a best overall response of stable disease. These data support further clinical development of capmatinib.
Collapse
Affiliation(s)
- Taito Esaki
- National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Fumihiko Hirai
- National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Akitaka Makiyama
- National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Takashi Seto
- National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Hideaki Bando
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoichi Naito
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | | | - Andrea Myers
- Novartis Institute for Biomedical Research, Shanghai, China
| | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
162
|
Puccini A, Marín-Ramos NI, Bergamo F, Schirripa M, Lonardi S, Lenz HJ, Loupakis F, Battaglin F. Safety and Tolerability of c-MET Inhibitors in Cancer. Drug Saf 2019; 42:211-233. [PMID: 30649748 PMCID: PMC7491978 DOI: 10.1007/s40264-018-0780-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The role of aberrant hepatocyte growth factor receptor (c-MET, also known as tyrosine-protein kinase MET)/hepatocyte growth factor (HGF) signaling in cancer progression and invasion has been extensively studied. c-MET inhibitors have shown promising pre-clinical and early phase clinical trial anti-tumor activity in several tumor types, although results of most phase III trials with these agents have been negative. To date, two small molecule c-MET inhibitors, cabozantinib and crizotinib, have been approved by regulatory authorities for the treatment of selected cancer types, but several novel c-MET inhibitors (either monoclonal antibodies or small molecule c-MET tyrosine kinase inhibitors) and treatment combinations are currently under study in different settings. Here we provide an overview of the mechanism of action and rationale of c-MET inhibition in cancer, the efficacy of approved agents, and novel promising c-MET-inhibitors and novel targeted combination strategies under development in different cancer types, with a focus on the safety profile and tolerability of these compounds.
Collapse
Affiliation(s)
- Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA, 90033, USA
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nagore I Marín-Ramos
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Francesca Bergamo
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marta Schirripa
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Sara Lonardi
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA, 90033, USA
| | - Fotios Loupakis
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA, 90033, USA.
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| |
Collapse
|
163
|
Mottaghitalab F, Farokhi M, Fatahi Y, Atyabi F, Dinarvand R. New insights into designing hybrid nanoparticles for lung cancer: Diagnosis and treatment. J Control Release 2019; 295:250-267. [DOI: 10.1016/j.jconrel.2019.01.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/22/2022]
|
164
|
Orlando E, Aebersold DM, Medová M, Zimmer Y. Oncogene addiction as a foundation of targeted cancer therapy: The paradigm of the MET receptor tyrosine kinase. Cancer Lett 2019; 443:189-202. [DOI: 10.1016/j.canlet.2018.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/20/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022]
|
165
|
Lee SH, Song J, Cho B, Hong S, Hoxha O, Kang T, Kim D, Lee LP. Bubble-free rapid microfluidic PCR. Biosens Bioelectron 2019; 126:725-733. [DOI: 10.1016/j.bios.2018.10.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 01/30/2023]
|
166
|
Abstract
The treatment landscape for non-small-cell lung cancer (NSCLC) has dramatically shifted over the past two decades. Targeted or precision medicine has primarily been responsible for this shift. Older paradigms of treating metastatic NSCLC with cytotoxic chemotherapy, while still important, have given way to evaluating tumor tissues for specific driver mutations that can be treated with targeted agents. Patients treated with targeted agents frequently have improved progression-free survival and overall survival compared to patients without a targetable driver mutation, highlighting the clinical benefit of precision medicine. In this chapter, we explore the historic landmark trials, the current state of the field, and potential future targets under investigation, in this exciting, rapidly evolving discipline of precision medicine in lung cancer.
Collapse
|
167
|
Metro G, Baglivo S, Siggillino A, Ludovini V, Chiari R, Rebonato A, Bellezza G. Successful Response to Osimertinib Rechallenge after Intervening Chemotherapy in an EGFR T790M-Positive Lung Cancer Patient. Clin Drug Investig 2018; 38:983-987. [PMID: 30151614 DOI: 10.1007/s40261-018-0691-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Osimertinib is the best treatment choice for patients with epidermal growth factor receptor (EGFR)-mutated advanced non-small-cell lung cancer (NSCLC) whose disease progresses on a first- or second-generation EGFR-tyrosine kinase inhibitor due to acquired T790M mutation. On the other hand, there is a lack of therapeutic strategies with proven efficacy at the time of progression on osimertinib. If not administered previously, platinum-based chemotherapy can provide some clinical benefit, while immunotherapy does not seem to work in this setting. Here, we report on a unique case of response to osimertinib rechallenge after intervening chemotherapy in an EGFR T790M-positive NSCLC patient pretreated with the sequence erlotinib-osimertinib.
Collapse
Affiliation(s)
- Giulio Metro
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Piazzale Menghini, 1, 06129, Perugia, Italy.
| | - Sara Baglivo
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Piazzale Menghini, 1, 06129, Perugia, Italy
| | - Annamaria Siggillino
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Piazzale Menghini, 1, 06129, Perugia, Italy
| | - Vienna Ludovini
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Piazzale Menghini, 1, 06129, Perugia, Italy
| | - Rita Chiari
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Piazzale Menghini, 1, 06129, Perugia, Italy
| | - Alberto Rebonato
- Department of Diagnostic Imaging, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Anatomic Pathology and Histology Section, Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
168
|
Crizotinib in advanced non-small-cell lung cancer with concomitant ALK rearrangement and c-Met overexpression. BMC Cancer 2018; 18:1171. [PMID: 30477470 PMCID: PMC6258481 DOI: 10.1186/s12885-018-5078-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023] Open
Abstract
Objective Crizotinib can target against mesenchymal-epithelial transition (MET) and anaplastic lymphoma kinase (ALK), which has been considered as a multi-targeted tyrosine kinase inhibitor (TKI). The objective of this study was to explore the efficacy of crizotinib in advanced non-small-cell lung cancer (NSCLC) with concomitant ALK rearrangement and c-Met overexpression. Methods Totally, 4622 advanced NSCLC patients from two institutes (3762 patients at the Guangdong Lung Cancer Institute from January 2011 to December 2016 and 860 cases at the Perking Cancer Hospital from January 2015 to December 2016) were screened for ALK rearrangement with any method of IHC, RACE-coupled PCR or FISH. C-Met expression was performed by IHC in ALK-rearranged patients, and more than 50% of cells with high staining were defined as c-Met overexpression. The efficacy of crizotinib was explored in the ALK-rearranged patients with or without c-Met overexpression. Results Sixteen patients were identified with c-Met overexpression in 160 ALK-rearranged cases, with the incidence of 10.0% (16/160). A total of 116 ALK-rearranged patients received the treatment of crizotinib. Objective response rate (ORR) was 86.7% (13/15) in ALK-rearranged patients with c-Met overexpression and 59.4% (60/101)in those without c-Met overexpression, P = 0.041. Median PFS showed a trend of superiority in c-Met overexpression group (15.2 versus 11.0 months, P = 0.263). Median overall survival (OS) showed a significant difference for ALK-rearranged patients with c-Met overexpression group of 33.5 months with the hazard ratio (HR) of 3.2. Conclusions C-Met overexpression co-exists with ALK rearrangement in a small population of advanced NSCLC. There may be a trend of favorable efficacy of crizotinib in such co-altered patients.
Collapse
|
169
|
Kim EK, Kim KA, Lee CY, Kim S, Chang S, Cho BC, Shim HS. Molecular Diagnostic Assays and Clinicopathologic Implications of MET Exon 14 Skipping Mutation in Non-small-cell Lung Cancer. Clin Lung Cancer 2018; 20:e123-e132. [PMID: 30391211 DOI: 10.1016/j.cllc.2018.10.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/27/2018] [Accepted: 10/02/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent studies revealed MET exon 14 skipping (METex14) as a biomarker that predicts the response to MET inhibitors in non-small-cell lung cancer (NSCLC). However, METex14 genomic alterations exhibit a highly diverse sequence composition, posing a challenge for clinical diagnostic testing. This study aimed to find a reasonable diagnostic assay for METex14 and identify its clinicopathologic implications. MATERIALS AND METHODS We performed a comprehensive analysis of METex14 in 414 EGFR/KRAS/ALK/ROS1-negative (quadruple negative) surgically resected NSCLCs. We used real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Sanger sequencing for the first assay, followed by next-generation sequencing (NGS; hybrid-capture targeted DNA/RNA sequencing). Clinicopathologic implications of the METex14 group were analyzed in a total of 880 NSCLCs. RESULTS METex14 was confirmed in 13 (3.1%) patients by DNA- and RNA-NGS. After comparison of assay results, qRT-PCR and NGS demonstrated the highest concordance rate. The mean variant allele frequency was 10.5% and 49% in DNA- and RNA-NGS, respectively. DNA-NGS revealed various lengths of indel and substitutions around and in exon 14. Moreover, METex14 was associated with adenocarcinoma (4.8%; 11/230) or sarcomatoid carcinoma (9.5%; 2/21), old age, never-smokers, and early stage of disease. CONCLUSIONS METex14 occurs in about 3% of NSCLCs and has characteristic clinicopathologic features. NGS should be the first assay of choice as a multiplex testing. Sanger sequencing can detect METex14, but sensitivity can be hampered by large deletions or low allele frequency. qRT-PCR, an mRNA-based method, is sensitive and specific and can be appropriate for screening METex14 as a single gene testing.
Collapse
Affiliation(s)
- Eun Kyung Kim
- Department of Pathology, National Health Insurance Service Ilsan Hospital, Goyang, Gyeonggi, Republic of Korea
| | - Kyung A Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang Young Lee
- Department of Cardiovascular and Thoracic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sangwoo Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sunhee Chang
- Department of Pathology, Ilsan Paik Hospital, Inje University, Goyang, Gyeonggi, Republic of Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
170
|
|
171
|
Mignard X, Ruppert AM, Antoine M, Vasseur J, Girard N, Mazières J, Moro-Sibilot D, Fallet V, Rabbe N, Thivolet-Bejui F, Rouquette I, Lantuejoul S, Cortot A, Saffroy R, Cadranel J, Lemoine A, Wislez M. c-MET Overexpression as a Poor Predictor of MET Amplifications or Exon 14 Mutations in Lung Sarcomatoid Carcinomas. J Thorac Oncol 2018; 13:1962-1967. [PMID: 30149144 DOI: 10.1016/j.jtho.2018.08.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/01/2018] [Accepted: 08/10/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION MNNG HOS transforming gene (MET) abnormalities such as amplification and exon 14 mutations may be responsive to targeted therapies. They are prevalent in lung sarcomatoid carcinomas (LSCs) and must be diagnosed as efficiently as possible. Hypothetically, c-MET overexpression by immunohistochemistry (IHC) may prove effective as a screening test for MET abnormalities. METHODS Tissue samples were obtained from consecutive patients with a resected LSC in four oncologic centers. IHC was performed using the SP44 antibody (Ventana, Tucson, Arizona) and evaluated using the MetMab score and H-score. Fluorescence in situ hybridization was applied with the dual color probe set from Zytovision (Clinisciences, Nanterre, France). True MET amplification was diagnosed when MET gene copy number was 5 or greater and the ratio between MET gene copy number and chromosome 7 number was greater than 2. All MET exon 14 alterations including those affecting splice sites occurring within splice donor and acceptor sites were detected in the routine molecular testing on genetic platforms. RESULTS A total of 81 LSCs were included. Fourteen (17%) exhibited positive IHC using the MetMab score and 15 (18.5%) using the H-score. MET amplification was detected in six tumors (8.5%) and MET exon 14 mutation in five (6%). A weak positive correlation between IHC and fluorescence in situ hybridization was found (r = 0.27, p = 0.0001). IHC sensitivity for MET amplification was 50%, with a specificity of 83%, positive predictive value of 21.4%, and negative predictive value of 94.7%. IHC sensitivity for MET exon 14 mutations was 20%, with a specificity of 83%, positive predictive value of 7%, and negative predictive value of 94%. CONCLUSION IHC is not a relevant screening tool for MET abnormalities in LSC.
Collapse
Affiliation(s)
- Xavier Mignard
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France
| | - Anne-Marie Ruppert
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | - Martine Antoine
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Cytology and Pathology Department, F-75970, Paris, France
| | - Julie Vasseur
- AP-HP, GH Paris-Sud, Paul Brousse Hospital, Department of Biochiemistry and Oncogenetics, Platform Oncomolpath/INCa, Villejuif, France
| | - Nicolas Girard
- Respiratory Medicine and Thoracic Oncology Service, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | | | | | - Vincent Fallet
- AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | - Nathalie Rabbe
- AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | | | - Isabelle Rouquette
- Toulouse Universitary Cancer Institute, IUCT-Oncopole, Pathology Department, Toulouse, France
| | - Sylvie Lantuejoul
- Biopathology Department, Léon Bérard Center, Unicancer Grenoble, France; Grenoble Alpes University, Grenoble, France
| | - Alexis Cortot
- CHU Lille, Thoracic Oncology Department, Univ. Lille, Siric ONCOLille, Lille, France
| | - Raphaël Saffroy
- AP-HP, GH Paris-Sud, Paul Brousse Hospital, Department of Biochiemistry and Oncogenetics, Platform Oncomolpath/INCa, Villejuif, France
| | - Jacques Cadranel
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | - Antoinette Lemoine
- AP-HP, GH Paris-Sud, Paul Brousse Hospital, Department of Biochiemistry and Oncogenetics, Platform Oncomolpath/INCa, Villejuif, France
| | - Marie Wislez
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France.
| |
Collapse
|
172
|
Wang M, Liang L, Lei X, Multani A, Meric-Bernstam F, Tripathy D, Wu Y, Chen H, Zhang H. Evaluation of cMET aberration by immunohistochemistry and fluorescence in situ hybridization (FISH) in triple negative breast cancers. Ann Diagn Pathol 2018; 35:69-76. [DOI: 10.1016/j.anndiagpath.2018.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
|
173
|
Su S, Dong ZY, Xie Z, Yan LX, Li YF, Su J, Liu SY, Yin K, Chen RL, Huang SM, Chen ZH, Yang JJ, Tu HY, Zhou Q, Zhong WZ, Zhang XC, Wu YL. Strong Programmed Death Ligand 1 Expression Predicts Poor Response and De Novo Resistance to EGFR Tyrosine Kinase Inhibitors Among NSCLC Patients With EGFR Mutation. J Thorac Oncol 2018; 13:1668-1675. [PMID: 30056164 DOI: 10.1016/j.jtho.2018.07.016] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 06/24/2018] [Accepted: 07/02/2018] [Indexed: 01/09/2023]
Abstract
INTRODUCTION This study evaluated whether tumor expression of programmed death ligand 1 (PD-L1) could predict the response of EGFR-mutated NSCLC to EGFR tyrosine kinase inhibitor (TKI) therapy. METHODS We retrospectively evaluated patients who received EGFR-TKIs for advanced NSCLC at the Guangdong Lung Cancer Institute between April 2016 and September 2017 and were not enrolled in clinical studies. The patients' EGFR and PD-L1 statuses were simultaneously evaluated. RESULTS Among the 101 eligible patients, strong PD-L1 expression significantly decreased objective response rate, compared with weak or negative PD-L1 expression (35.7% versus 63.2% versus 67.3%, p = 0.002), and shortened progression-free survival (3.8 versus 6.0 versus 9.5 months, p < 0.001), regardless of EGFR mutation type (19del or L858R). Furthermore, positive PD-L1 expression was predominantly observed among patients with de novo resistance rather than acquired resistance to EGFR-TKIs (66.7% versus 30.2%, p = 0.009). Notably, we found a high proportion of PD-L1 and cluster of differentiation 8 (CD8) dual-positive cases among patients with de novo resistance (46.7%, 7 of 15). Finally, one patient with de novo resistance to EGFR-TKIs and PD-L1 and CD8 dual positivity experienced a favorable response to anti-programmed death 1 therapy. CONCLUSIONS This study revealed the adverse effects of PD-L1 expression on EGFR-TKI efficacy, especially in NSCLC patients with de novo resistance. The findings indicate the reshaping of an inflamed immune phenotype characterized by PD-L1 and CD8 dual positivity and suggest potential therapeutic sensitivity to programmed death 1 blockade.
Collapse
Affiliation(s)
- Shan Su
- Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhong-Yi Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Li-Xu Yan
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu-Fa Li
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kai Yin
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Rui-Lian Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shu-Mei Huang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi-Long Wu
- Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
174
|
Zhang M, Li G, Sun X, Ni S, Tan C, Xu M, Huang D, Ren F, Li D, Wei P, Du X. MET amplification, expression, and exon 14 mutations in colorectal adenocarcinoma. Hum Pathol 2018; 77:108-115. [PMID: 29641976 DOI: 10.1016/j.humpath.2018.03.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 02/07/2023]
Abstract
MET amplification, expression, and splice mutations at exon 14 result in dysregulation of the MET signaling pathway. The aim of this study was to identify the relationship between MET amplification, protein or mRNA expression, and mutations in colorectal cancer (CRC). MET immunohistochemistry was used for MET protein expression analysis, and fluorescence in situ hybridization was used for MET amplification detection. Both analyses were performed in tissue microarrays containing 294 colorectal adenocarcinoma tissue samples and 131 samples of adjacent normal epithelial tissue. MET mRNA expression was examined by real-time quantitative polymerase chain reaction in 72 fresh colorectal adenocarcinoma tissue samples and adjacent normal colon tissue. Polymerase chain reaction sequencing was performed to screen for MET exon 14 splice mutations in 59 fresh CRC tissue samples. Our results showed that MET protein expression was higher in colorectal tumor tissue than in adjacent normal intestinal epithelium. Positive MET protein expression was associated with significantly poorer overall survival and disease-free survival. Multivariate analysis revealed that positive MET protein expression was an independent risk factor for disease-free survival but not for overall survival. MET mRNA expression was upregulated in tumor tissues compared with the adjacent normal tissues. The incidence of MET amplification was 4.4%. None of the patients was positive for MET mutation. Collectively, MET was overexpressed in colorectal adenocarcinoma, and its positive protein expression predicted a poorer outcome in CRC patients. Furthermore, according to our results, MET amplification and exon 14 mutation are extremely rare events in colorectal adenocarcinoma.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China.
| | - Guichao Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Xiangjie Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Shujuan Ni
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Fei Ren
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Dawei Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Institute of Pathology, Fudan University, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
175
|
Rieber N, Bohnert R, Ziehm U, Jansen G. Reliability of algorithmic somatic copy number alteration detection from targeted capture data. Bioinformatics 2018; 33:2791-2798. [PMID: 28472276 PMCID: PMC5870863 DOI: 10.1093/bioinformatics/btx284] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/03/2017] [Indexed: 01/11/2023] Open
Abstract
Motivation Whole exome and gene panel sequencing are increasingly used for oncological diagnostics. To investigate the accuracy of SCNA detection algorithms on simulated and clinical tumor samples, the precision and sensitivity of four SCNA callers were measured using 50 simulated whole exome and 50 simulated targeted gene panel datasets, and using 119 TCGA tumor samples for which SNP array data were available. Results On synthetic exome and panel data, VarScan2 mostly called false positives, whereas Control-FREEC was precise (>90% correct calls) at the cost of low sensitivity (<40% detected). ONCOCNV was slightly less precise on gene panel data, with similarly low sensitivity. This could be explained by low sensitivity for amplifications and high precision for deletions. Surprisingly, these results were not strongly affected by moderate tumor impurities; only contaminations with more than 60% non-cancerous cells resulted in strongly declining precision and sensitivity. On the 119 clinical samples, both Control-FREEC and CNVkit called 71.8% and 94%, respectively, of the SCNAs found by the SNP arrays, but with a considerable amount of false positives (precision 29% and 4.9%). Discussion Whole exome and targeted gene panel methods by design limit the precision of SCNA callers, making them prone to false positives. SCNA calls cannot easily be integrated in clinical pipelines that use data from targeted capture-based sequencing. If used at all, they need to be cross-validated using orthogonal methods. Availability and implementation Scripts are provided as supplementary information. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nora Rieber
- Molecular Health GmbH, Kurfürsten-Anlage 21, 69115 Heidelberg, Germany
| | - Regina Bohnert
- Molecular Health GmbH, Kurfürsten-Anlage 21, 69115 Heidelberg, Germany
| | - Ulrike Ziehm
- Molecular Health GmbH, Kurfürsten-Anlage 21, 69115 Heidelberg, Germany
| | - Gunther Jansen
- Molecular Health GmbH, Kurfürsten-Anlage 21, 69115 Heidelberg, Germany
| |
Collapse
|
176
|
Davies KD, Le AT, Sheren J, Nijmeh H, Gowan K, Jones KL, Varella-Garcia M, Aisner DL, Doebele RC. Comparison of Molecular Testing Modalities for Detection of ROS1 Rearrangements in a Cohort of Positive Patient Samples. J Thorac Oncol 2018; 13:1474-1482. [PMID: 29935306 DOI: 10.1016/j.jtho.2018.05.041] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/25/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022]
Abstract
INTRODUCTION ROS1 gene fusions are a well-characterized class of oncogenic driver found in approximately 1% to 2% of NSCLC patients. ROS1-directed therapy in these patients is more efficacious and is associated with fewer side effects compared to chemotherapy and is thus now considered standard-of-care for patients with advanced disease. Consequently, accurate detection of ROS1 rearrangements/fusions in clinical tumor samples is vital. In this study, we compared the performance of three common molecular testing approaches on a cohort of ROS1 rearrangement/fusion-positive patient samples. METHODS Twenty-three ROS1 rearrangement/fusion-positive clinical samples were assessed by at least two of the following molecular testing methodologies: break-apart fluorescence in situ hybridization, DNA-based hybrid capture library preparation followed by next-generation sequencing (NGS), and RNA-based anchored multiplex polymerase chain reaction library preparation followed by NGS. RESULTS None of the testing methodologies demonstrated 100% sensitivity in detection of ROS1 rearrangements/fusions. Fluorescence in situ hybridization results were negative in 2 of 20 tested samples, the DNA-based NGS assay was negative in 4 of 18 tested samples, and the RNA-based NGS assay was negative in 3 of 19 tested samples. For all three testing approaches, we identified assay characteristics that likely contributed to false-negative results. Additionally, we report that genomic breakpoints are an unreliable predictor of breakpoints at the transcript level, likely due to alternative splicing. CONCLUSIONS ROS1 rearrangement/fusion detection in the clinical setting is complex and all methodologies have inherent limitations of which users must be aware to correctly interpret results.
Collapse
Affiliation(s)
- Kurtis D Davies
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Anh T Le
- Department of Medicine - Division of Medical Oncology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Jamie Sheren
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Hala Nijmeh
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Katherine Gowan
- Department of Pediatrics - Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Kenneth L Jones
- Department of Pediatrics - Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Marileila Varella-Garcia
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado; Department of Medicine - Division of Medical Oncology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado
| | - Robert C Doebele
- Department of Medicine - Division of Medical Oncology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
177
|
Scagliotti GV, Shuster D, Orlov S, von Pawel J, Shepherd FA, Ross JS, Wang Q, Schwartz B, Akerley W. Tivantinib in Combination with Erlotinib versus Erlotinib Alone for EGFR-Mutant NSCLC: An Exploratory Analysis of the Phase 3 MARQUEE Study. J Thorac Oncol 2018; 13:849-854. [DOI: 10.1016/j.jtho.2017.12.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 01/22/2023]
|
178
|
Affiliation(s)
- Alex Dmitrienko
- From Mediana, Overland Park, KS (A.D.); and Boston University, Boston (R.B.D.)
| | - Ralph B D'Agostino
- From Mediana, Overland Park, KS (A.D.); and Boston University, Boston (R.B.D.)
| |
Collapse
|
179
|
Mauri G, Valtorta E, Cerea G, Amatu A, Schirru M, Marrapese G, Fiorillo V, Recchimuzzo P, Cavenago IS, Bonazzina EF, Motta V, Lauricella C, Veronese S, Tosi F, Maiolani M, Rospo G, Truini M, Bonoldi E, Christiansen J, Potts SJ, Siena S, Sartore-Bianchi A. TRKA expression and NTRK1 gene copy number across solid tumours. J Clin Pathol 2018; 71:926-931. [PMID: 29802225 DOI: 10.1136/jclinpath-2018-205124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/18/2018] [Accepted: 05/04/2018] [Indexed: 12/17/2022]
Abstract
AIMS Neurotrophic Tropomyosin Kinase Receptor 1 (NTRK1) gene encodes for the protein Tropomyosin-related kinase A (TRKA). Deregulated activity of TRKA has been shown to have oncogenic potential. We present here the results of an immunohistochemical (IHC) observational cohort study of TRKA expression together with gene copy number (GCN) assessment in various solid tumours. METHODS Formalin-fixed, paraffin-embedded consecutive samples of different tumour types were tested for TRKA expression. Samples showing TRKA IHC staining in at least 10% of cells were analysed by fluorescence in situ hybridisation to assess NTRK1 gene rearrangements and/or individual GCN gain. All patients underwent this molecular assessment within the phase I ALKA-001 clinical trial. RESULTS 1043 samples were tested and annotation for histology was available in 1023. Most of the samples were colorectal adenocarcinoma (CRC) (n=550, 52.7%) and lung adenocarcinoma (n=312, 29.9%). 24 samples (2.3%) were biliary tract carcinoma (BTC). Overall, 17 (1.6%) samples were characterised by TRKA IHC expression (four weak, eight moderate, five strong): 9/17 lung adenocarcinoma, 3/17 CRC, 3/17 BTC, 1/17 thyroid cancer and 1/17 cancer of unknown primary. Of these, 1/17 with strong TRKA IHC staining displayed NTRK1 gene rearrangement and 15/17 NTRK1 GCN gain by FISH. No correlation was found between intensity of TRKA IHC staining and number of copies of NTRK1. CONCLUSIONS TRKA expression can be found in 1.6% of solid tumours and can be paralleled by NTRK1 gene rearrangements or mostly GCN gain. The prognostic and translational therapeutic impact of the latter remains to be established.
Collapse
Affiliation(s)
- Gianluca Mauri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Ematologia e Onco-Ematologia, Università degli Studi di Milano, Milan, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giulio Cerea
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Michele Schirru
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giovanna Marrapese
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Vincenzo Fiorillo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Patrizia Recchimuzzo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Ivana Stella Cavenago
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Valentina Motta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Calogero Lauricella
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Silvio Veronese
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Ematologia e Onco-Ematologia, Università degli Studi di Milano, Milan, Italy
| | - Martina Maiolani
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Ematologia e Onco-Ematologia, Università degli Studi di Milano, Milan, Italy
| | | | - Mauro Truini
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Emanuela Bonoldi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Laboratory Medicine, Division of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | | | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Ematologia e Onco-Ematologia, Università degli Studi di Milano, Milan, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Ematologia e Onco-Ematologia, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
180
|
Yen YC, Hsu HL, Chang JH, Lin WC, Chang YC, Chang CL, Chow JM, Yuan KSP, Wu ATH, Wu SY. Efficacy of thoracic radiotherapy in patients with stage IIIB-IV epidermal growth factor receptor-mutant lung adenocarcinomas who received and responded to tyrosine kinase inhibitor treatment. Radiother Oncol 2018; 129:52-60. [PMID: 29703499 DOI: 10.1016/j.radonc.2018.03.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 02/05/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Large-scale, prospective, randomized studies of the efficacy of thoracic radiotherapy (RT) in patients with unresectable stage IIIB-IV epidermal growth factor receptor (EGFR)-mutant lung adenocarcinomas who received and responded to EGFR tyrosine kinase inhibitor (TKI) treatment are not currently available. Therefore, we designed a propensity score-matched, nationwide, population-based, cohort study for estimating the effects of thoracic RT on patients with EGFR-mutant lung adenocarcinomas. PATIENTS AND METHODS We analyzed patients with unresectable stage IIIB-IV EGFR mutant lung adenocarcinomas and categorized them into two groups according to treatment modality and compared their outcomes; groups 1 and 2 consisted of patients who received EGFR TKI treatment alone until tumor progression and those who received and responded to EGFR TKI treatment and subsequently received thoracic RT for lung tumors, respectively. The patients in groups 2 and 1 were matched at a ratio of 1:4. RESULTS The matching process yielded a final cohort of 1475 patients (1180 and 295 patients in groups 1 and 2, respectively) who were eligible for further analysis. According to both univariate and multivariate Cox regression analyses, the adjusted hazard ratios (aHRs) (95% confidence interval [CI]) derived for thoracic RT for lung tumor after EGFR TKI use and tumor response (group 2) compared with EGFR TKI treatment alone (group 1) was 0.72 (0.60-0.85). CONCLUSIONS Thoracic RT might be associated with overall survival in patients with unresectable stage IIIB-IV EGFR-mutant lung adenocarcinomas who received and responded to EGFR TKI treatment.
Collapse
Affiliation(s)
- Yu-Chun Yen
- Biostatistics Center and School of Public Health, Taipei Medical University, Taiwan
| | - Han-Lin Hsu
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taiwan
| | - Jer-Hwa Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taiwan
| | - Wei-Cheng Lin
- Division of Thoracic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Yin-Chun Chang
- Division of Thoracic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Chia-Lun Chang
- Department of Hematology and Oncology, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Jyh-Ming Chow
- Department of Hematology and Oncology, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Kevin Sheng-Po Yuan
- Department of Otorhinolaryngology, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Alexander T H Wu
- Ph.D. Program for Translational Medicine, Taipei Medical University, Taiwan
| | - Szu-Yuan Wu
- Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taiwan.
| |
Collapse
|
181
|
Kim JH, Kim HS, Kim BJ. Prognostic value of MET copy number gain in non-small-cell lung cancer: an updated meta-analysis. J Cancer 2018; 9:1836-1845. [PMID: 29805710 PMCID: PMC5968772 DOI: 10.7150/jca.24980] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/17/2018] [Indexed: 12/14/2022] Open
Abstract
The alterations of MET have been detected in non-small-cell lung cancer (NSCLC). However, the prognostic impact of MET gene copy number gain (CNG) has not been consistent among studies. We performed this meta-analysis to evaluate the prognostic value of high MET CNG in patients with NSCLC. A systematic computerized search of the electronic databases including PubMed, EMBASE, Google scholar, and Cochrane Library (up to November 2017) was carried out. From twenty-one studies, 7,647 patients were included in the pooled analysis of hazard ratios (HRs) with 95% confidence intervals (CIs) for disease-free survival or overall survival. Compared with patients with NSCLC showing low MET CNG, those with tumors harboring high MET CNG showed significantly worse survival (HR = 1.45, 95% CI: 1.16-1.80, p = 0.001). Subgroup analyses showed that high MET CNG significantly correlated with a poor prognosis especially in patients with adenocarcinoma (HR = 1.41, 95% CI: 1.11-1.79, p = 0.005) and Asian populations (HR = 1.58, 95% CI: 1.32-1.88, p < 0.00001). In conclusion, this meta-analysis indicates that high MET CNG is an adverse prognostic factor in patients with NSCLC. Subgroup analyses suggest that high MET CNG is associated with a worse prognosis, especially in patients with adenocarcinoma and Asian populations. However, large prospective studies using standardized methods based on the homogeneous populations are warranted to validate the prognostic value of MET amplification in patients with 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, 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, 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, Republic of Korea.,Department of Internal Medicine, National Army Capital Hospital, The Armed Forces Medical Command, Sungnam, Gyeonggi-Do, Republic of Korea
| |
Collapse
|
182
|
Tu CY, Cheng FJ, Chen CM, Wang SL, Hsiao YC, Chen CH, Hsia TC, He YH, Wang BW, Hsieh IS, Yeh YL, Tang CH, Chen YJ, Huang WC. Cigarette smoke enhances oncogene addiction to c-MET and desensitizes EGFR-expressing non-small cell lung cancer to EGFR TKIs. Mol Oncol 2018; 12:705-723. [PMID: 29570930 PMCID: PMC5928373 DOI: 10.1002/1878-0261.12193] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/13/2018] [Accepted: 02/20/2018] [Indexed: 12/23/2022] Open
Abstract
Cigarette smoking is one of the leading risks for lung cancer and is associated with the insensitivity of non‐small cell lung cancer (NSCLC) to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). However, it remains undetermined whether and how cigarette smoke affects the therapeutic efficacy of EGFR TKIs. In this study, our data showed that chronic exposure to cigarette smoke extract (CSE) or tobacco smoke‐derived carcinogen benzo[α]pyrene, B[α]P, but not nicotine‐derived nitrosamine ketone (NNK), reduced the sensitivity of wild‐type EGFR‐expressing NSCLC cells to EGFR TKIs. Treatment with TKIs almost abolished EGFR tyrosine kinase activity but did not show an inhibitory effect on downstream Akt and ERK pathways in B[α]P‐treated NSCLC cells. CSE and B[α]P transcriptionally upregulate c‐MET and activate its downstream Akt pathway, which is not inhibited by EGFR TKIs. Silencing of c‐MET reduces B[α]P‐induced Akt activation. The CSE‐treated NSCLC cells are sensitive to the c‐MET inhibitor crizotinib. These findings suggest that cigarette smoke augments oncogene addiction to c‐MET in NSCLC cells and that MET inhibitors may show clinical benefits for lung cancer patients with a smoking history.
Collapse
Affiliation(s)
- Chih-Yen Tu
- Department of Life Science, the iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan
| | - Fang-Ju Cheng
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chuan-Mu Chen
- Department of Life Science, the iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Ling Wang
- Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| | - Yu-Chun Hsiao
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Chia-Hung Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of Respiratory Therapy, China Medical University, Taichung, Taiwan
| | - Te-Chun Hsia
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of Respiratory Therapy, China Medical University, Taichung, Taiwan.,Hyperbaric Oxygen Therapy Center, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Hao He
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Bo-Wei Wang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - I-Shan Hsieh
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Yi-Lun Yeh
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Yun-Ju Chen
- Department of Medical Research, E-DA Hospital, Kaohsiung, Taiwan.,Department of Biological Science & Technology, I-Shou University, Kaohsiung, Taiwan.,School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Wei-Chien Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan.,The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan.,Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, China Medical University and Hospital, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.,Research Center for New Drug Development, China Medical University, Taichung, Taiwan
| |
Collapse
|
183
|
McCoach CE, Le AT, Gowan K, Jones K, Schubert L, Doak A, Estrada-Bernal A, Davies KD, Merrick DT, Bunn PA, Purcell WT, Dziadziuszko R, Varella-Garcia M, Aisner DL, Camidge DR, Doebele RC. Resistance Mechanisms to Targeted Therapies in ROS1+ and ALK+ Non-small Cell Lung Cancer. Clin Cancer Res 2018; 24:3334-3347. [PMID: 29636358 DOI: 10.1158/1078-0432.ccr-17-2452] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 02/23/2018] [Accepted: 04/03/2018] [Indexed: 12/13/2022]
Abstract
Purpose: Despite initial benefit from tyrosine kinase inhibitors (TKIs), patients with advanced non-small cell lung cancer (NSCLC) harboring ALK (ALK+) and ROS1 (ROS1+) gene fusions ultimately progress. Here, we report on the potential resistance mechanisms in a series of patients with ALK+ and ROS1+ NSCLC progressing on different types and/or lines of ROS1/ALK-targeted therapy.Experimental Design: We used a combination of next-generation sequencing (NGS), multiplex mutation assay, direct DNA sequencing, RT-PCR, and FISH to identify fusion variants/partners and copy-number gain (CNG), kinase domain mutations (KDM), and copy-number variations (CNVs) in other cancer-related genes. We performed testing on 12 ROS1+ and 43 ALK+ patients.Results: One of 12 ROS1+ (8%) and 15 of 43 (35%) ALK + patients harbored KDM. In the ROS1+ cohort, we identified KIT and β-catenin mutations and HER2-mediated bypass signaling as non-ROS1-dominant resistance mechanisms. In the ALK+ cohort, we identified a novel NRG1 gene fusion, a RET fusion, 2 EGFR, and 3 KRAS mutations, as well as mutations in IDH1, RIT1, NOTCH, and NF1 In addition, we identified CNV in multiple proto-oncogenes genes including PDGFRA, KIT, KDR, GNAS, K/HRAS, RET, NTRK1, MAP2K1, and others.Conclusions: We identified a putative TKI resistance mechanism in six of 12 (50%) ROS1 + patients and 37 of 43 (86%) ALK+ patients. Our data suggest that a focus on KDMs will miss most resistance mechanisms; broader gene testing strategies and functional validation is warranted to devise new therapeutic strategies for drug resistance. Clin Cancer Res; 24(14); 3334-47. ©2018 AACR.
Collapse
Affiliation(s)
- Caroline E McCoach
- Division of Medical Oncology, UCSF Helen Diller Comprehensive Cancer Center, San Francisco, California.
| | - Anh T Le
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine Gowan
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora, Colorado
| | - Kenneth Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora, Colorado
| | - Laura Schubert
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Andrea Doak
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Adriana Estrada-Bernal
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Kurtis D Davies
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel T Merrick
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Paul A Bunn
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - W Tom Purcell
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Marileila Varella-Garcia
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
184
|
MET exon 14 mutations as targets in routine molecular analysis of primary sarcomatoid carcinoma of the lung. Oncotarget 2018; 8:42428-42437. [PMID: 28418914 PMCID: PMC5522077 DOI: 10.18632/oncotarget.16403] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/20/2017] [Indexed: 11/25/2022] Open
Abstract
MET exon 14 splicing mutations are new targetable oncogenic drivers reported in 3% of non-small cell lung cancer (NSCLC) cases and have been shown to be more common in pulmonary sarcomatoid carcinomas (PSCs). This study sought to screen mutations affecting MET exon 14 splice sites in a large SC cohort of Caucasian patients, with a large adenocarcinoma cohort as internal control. We tested 81 patients with SC and 150 with adenocarcinoma for splice site DNA mutations leading to RNA splicing-based skipping of MET exon 14. To this end, we employed a mass spectrometry-based custom-designed PCR assay for routine analysis of whole MET exon 14 and flanking intronic regions using formalin-fixed paraffin-embedded (FFPE) tumor samples. Our results revealed a 4.9% mutation rate for MET exon 14 mutations in Caucasian SC patients, which is, though highly variable, within the usual range reported in NSCLC. Discrepancies with previous results reported in SC could be accounted for the small number of cases, ethnicity, epithelial component, and percentage of other driver mutations, such as KRAS, in the patient populations studied. Based on our study findings, SC patients should be screened for MET exon 14 mutations in the same manner as adenocarcinoma patients.
Collapse
|
185
|
Zhou Y, Zhang Z, Wang N, Chen J, Zhang X, Guo M, John Zhong L, Wang Q. Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial-mesenchymal transition in lung adenocarcinoma. Cell Death Dis 2018; 9:429. [PMID: 29559623 PMCID: PMC5861121 DOI: 10.1038/s41419-018-0457-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 01/30/2023]
Abstract
Non-small cell lung cancer (NSCLC), including adenocarcinoma and squamous cell carcinoma, is the leading cause of death from lung malignancies and has a poor prognosis due to metastasis. Suppressor of cytokine signalling-2 (SOCS2), a feedback inhibitor of cytokine signalling, has been shown to be involved in growth control. Here, we show that SOCS2 were significantly downregulated in tumour foci in NSCLC patients. The expression levels of SOCS2 significantly correlated with clinical stage, lymph node metastasis, histological subtype and survival time. In particular, the decreased expression of SOCS2 significantly associated with advanced pathological stage, lymph node metastasis and shorter overall survival in lung adenocarcinoma patients. In vivo animal results showed that overexpressed SOCS2 attenuated the metastatic characteristics of lung adenocarcinoma, including by inhibiting the epithelial-mesenchymal transition (EMT). Further functional studies indicated that insulin-like growth factor 1 (IGF1)-driven migratory and invasive behaviours of lung adenocarcinoma cells can be partially suppressed by exogenous SOCS2 expression. Investigations into the mechanism of action revealed that SOCS2 inhibits EMT by inactivating signal transducer and activator of transcription 3 (STAT3) and STAT5 via the competitive binding of SOCS2 to the STAT binding sites on IGF1R. Altogether, our results reveal an important role for SOCS2 dysregulation in the pathogenicity of lung adenocarcinoma, suggest its potential use as a biomarker for diagnosing lung adenocarcinoma, and paves the way to develop novel therapy targets as the axis of SOCS2-IGF1R-STAT in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Thoracic Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhilei Zhang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210029, China
| | - Ning Wang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210029, China
| | - Jizheng Chen
- State Key Lab of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xu Zhang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210029, China
| | - Min Guo
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Li John Zhong
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210029, China
| | - Qian Wang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
186
|
The multiple paths towards MET receptor addiction in cancer. Oncogene 2018; 37:3200-3215. [PMID: 29551767 DOI: 10.1038/s41388-018-0185-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/30/2018] [Accepted: 01/30/2018] [Indexed: 12/14/2022]
Abstract
Targeted therapies against receptor tyrosine kinases (RTKs) are currently used with success on a small proportion of patients displaying clear oncogene activation. Lung cancers with a mutated EGFR provide a good illustration. The efficacy of targeted treatments relies on oncogene addiction, a situation in which the growth or survival of the cancer cells depends on a single deregulated oncogene. MET, a member of the RTK family, is a promising target because it displays many deregulations in a broad panel of cancers. Although clinical trials having evaluated MET inhibitors in large populations have yielded disappointing results, many recent case reports suggest that MET inhibition may be effective in a subset of patients with unambiguous MET activation and thus, most probably, oncogene addiction. Interestingly, preclinical studies have revealed a particularity of MET addiction: it can arise through several mechanisms, and the mechanism involved can differ according to the cancer type. The present review describes the different mechanisms of MET addiction and their consequences for diagnosis and therapeutic strategies. Although in each cancer type MET addiction affects a restricted number of patients, pooling of these patients across all cancer types yields a targetable population liable to benefit from addiction-targeting therapies.
Collapse
|
187
|
Park CK, Oh IJ, Choi YD, Jang TW, Lee JE, Ryu JS, Lee SY, Kim YC. A Prospective Observational Study Evaluating the Correlation of c-MET Expression and EGFR Gene Mutation with Response to Erlotinib as Second-Line Treatment for Patients with Advanced/Metastatic Non-Small-Cell Lung Cancer. Oncology 2018; 94:373-382. [PMID: 29502124 DOI: 10.1159/000486896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/12/2018] [Indexed: 03/22/2025]
Abstract
OBJECTIVES We aimed to evaluate the prevalence and predictive role of c-MET expression and EGFR mutation in the efficacy of erlotinib in non-small-cell lung cancer (NSCLC). METHODS We prospectively recruited 196 patients with stage IV or recurrent NSCLC treated with erlotinib after failure of first-line chemotherapy. Immunohistochemistry was used to evaluate c-MET overexpression, silver in situ hybridization (SISH) to assess gene copy number, and real-time polymerase chain reaction to detect EGFR mutations, respectively, in tumor tissue. RESULTS The major histologic type was adenocarcinoma (66.8%). c-MET was overexpressed in 55.8% (87/156) and dominant in females as well as non-squamous histology. Although c-MET gene amplification and high polysomy were observed in 2.0% (3/152) and 11.2% (17/152), they did not correlate with any characteristics. EGFR mutation was detected in 13.1% (20/153). The objective response rate of erlotinib was higher (61.1 vs. 3.7%, p < 0.001) and the median progression-free survival (PFS) was longer (10.2 vs. 1.9 months, p < 0.001) in EGFR-sensitizing mutations. However, c-MET positivity did not show a significant correlation with response to erlotinib or PFS. CONCLUSION We reconfirmed EGFR mutation as a strong predictive marker of NSCLC. However, c-MET positivity was not associated with response or PFS, although c-MET overexpression correlated with some clinical characteristics.
Collapse
Affiliation(s)
- Cheol-Kyu Park
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Jeonnam, Republic of Korea
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Jeonnam, Republic of Korea,
| | - Yoo-Duk Choi
- Department of Pathology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Tae-Won Jang
- Department of Internal Medicine, Kosin University Gospel Hospital, Busan, Republic of Korea
| | - Jeong-Eun Lee
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jeong-Seon Ryu
- Department of Internal Medicine, Inha University Hospital, Incheon, Republic of Korea
| | - Shin-Yup Lee
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Young-Chul Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Jeonnam, Republic of Korea
| |
Collapse
|
188
|
Guo Y, Tao M, Jiang M. MicroRNA-454-3p inhibits cervical cancer cell invasion and migration by targeting c-Met. Exp Ther Med 2018; 15:2301-2306. [PMID: 29563976 PMCID: PMC5854935 DOI: 10.3892/etm.2018.5714] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 11/22/2017] [Indexed: 01/15/2023] Open
Abstract
Increasing evidence has demonstrated that microRNAs (miRNAs) have a crucial role in the initiation and progression of tumors. The present study aimed to investigate the expression and the role of miRNA-454-3p in human cervical cancer. Human cervical cancer cells were transfected with miRNA-454-3p mimics or negative control miRNA. MTT, Transwell and wound healing assays were performed to investigate the effects of miRNA-454-3p overexpression on cell proliferation, invasion and migration, respectively. The results indicated that miRNA-454-3p was down-regulated in human cervical cancer cell lines, while its ectopic overexpression significantly inhibited their proliferation, migration and invasion. Furthermore, a luciferase reporter assay confirmed that c-met was a novel target of miRNA-454-3p in HeLa cells. In conclusion, the results of the present study suggested that miRNA-454-3p exhibits significant tumor-suppressive effects in cervical cancer by targeting c-met, and may be a potential means of treating cervical cancer.
Collapse
Affiliation(s)
- Yan Guo
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Min Jiang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
189
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 2018; 142:321-346. [PMID: 29355391 DOI: 10.5858/arpa.2017-0388-cp] [Citation(s) in RCA: 573] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT - In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE - To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN - The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS - Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS - The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes ( ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- From the Departments of Pathology (Drs Lindeman and Sholl) and Medicine (Dr Kwiatkowski), Brigham and Women's Hospital, Boston, Massachusetts; the Cancer Center (Dr Bernicker) and the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas (Dr Cagle); the Department of Pathology, University of Colorado School of Medicine, Denver (Dr Aisner); the Diagnostic and Molecular Pathology Laboratory (Dr Arcila) and the Molecular Diagnostics Service (Dr Ladanyi), Memorial Sloan Kettering Cancer Center, New York, New York; the Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York (Dr Beasley); the Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois (Mss Colasacco and Ventura); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Medicine and Pathology, University of Colorado, Denver (Dr Hirsch); the Department of Pathology, University of Aberdeen, Aberdeen, Scotland (Dr Kerr); the Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland (Dr Temple-Smolkin); the Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia (Dr Solomon); the Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands (Dr Thunnissen); the Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada (Dr Tsao); Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado (Dr Wynes); and the Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan (Dr Yatabe). Dr Souter is in private practice in Wellanport, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Thorac Oncol 2018; 13:323-358. [PMID: 29396253 DOI: 10.1016/j.jtho.2017.12.001] [Citation(s) in RCA: 357] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 12/15/2022]
Abstract
CONTEXT In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes (ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Philip T Cagle
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Denver, New York
| | - Maria E Arcila
- Diagnostic and Molecular Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mary Beth Beasley
- Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York
| | | | - Carol Colasacco
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fred R Hirsch
- Department of Medicine and Pathology, University of Colorado, Denver, New York
| | - Keith Kerr
- Department of Pathology, University of Aberdeen, Aberdeen, Scotland
| | | | - Marc Ladanyi
- Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan A Nowak
- Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robyn Temple-Smolkin
- Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland
| | - Benjamin Solomon
- Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia
| | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Christina B Ventura
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Murry W Wynes
- Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
191
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Mol Diagn 2018; 20:129-159. [PMID: 29398453 DOI: 10.1016/j.jmoldx.2017.11.004] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
CONTEXT In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes (ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Philip T Cagle
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Denver, Colorado
| | - Maria E Arcila
- Diagnostic and Molecular Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mary Beth Beasley
- Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York
| | - Eric H Bernicker
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas
| | - Carol Colasacco
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fred R Hirsch
- Department of Medicine and Pathology, University of Colorado, Denver, Colorado
| | - Keith Kerr
- Department of Pathology, University of Aberdeen, Aberdeen, Scotland
| | | | - Marc Ladanyi
- Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan A Nowak
- Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robyn Temple-Smolkin
- Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland
| | - Benjamin Solomon
- Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia
| | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Christina B Ventura
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Murry W Wynes
- Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
192
|
Lou NN, Zhang XC, Chen HJ, Zhou Q, Yan LX, Xie Z, Su J, Chen ZH, Tu HY, Yan HH, Wang Z, Xu CR, Jiang BY, Wang BC, Bai XY, Zhong WZ, Wu YL, Yang JJ. Clinical outcomes of advanced non-small-cell lung cancer patients with EGFR mutation, ALK rearrangement and EGFR/ALK co-alterations. Oncotarget 2018; 7:65185-65195. [PMID: 27533086 PMCID: PMC5323147 DOI: 10.18632/oncotarget.11218] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
The co-occurrence of epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements constitutes a rare molecular subtype of non-small-cell lung cancer (NSCLC). Herein, we assessed the clinical outcomes and incidence of acquired resistance to tyrosine kinase inhibitors (TKIs) in this subtype. So we enrolled 118 advanced NSCLC treated with TKIs. EGFR mutations and ALK rearrangements were detected by DNA sequencing or Scorpion amplification refractory mutation system and fluorescence in situ hybridization respectively. Immunohistochemistry was used to evaluate the activation of associated proteins. We found that nine in ten patients with EGFR/ALK co-alterations had good response with first-line EGFR TKI, and the objective response rate (ORR) of EGFR TKIs was 80% (8/10) for EGFR/ALK co-altered and 65.5% (55/84) for EGFR-mutant (P = 0.57), with a median progression-free survival (PFS) of 11.2 and 13.2 months, (hazard ratio [HR]=0.95, 95% [CI], 0.49-1.84, P= 0.87). ORR of crizotinib was 40% (2/5) for EGFR/ALK co-altered and 73.9% (17/23) for ALK-rearranged (P= 0.29), with a median PFS of 1.9 and 6.9 months (hazard ratio [HR], 0.40; 95% [CI] 0.15-1.10, P = 0.08). The median overall survival (OS) was 21.3, 23.7, and 18.5 months in EGFR-mutant, ALK-rearranged, and EGFR/ALK co-altered (P= 0.06), and there existed a statistically significant difference in OS between ALK-rearranged and EGFR/ALK co-altered (P=0.03). Taken together, the first-line EGFR-TKI might be the reasonable care for advanced NSCLC harbouring EGFR/ALK co-alterations, whether or nor to use sequential crizotinib should be guided by the status of ALK rearrangement and the relative level of phospho-EGFR and phospho-ALK.
Collapse
Affiliation(s)
- Na-Na Lou
- Southern Medical University, Guangzhou, 510515, China.,Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.,Medical Research Center, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.,Medical Research Center, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hua-Jun Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Li-Xu Yan
- Department of Pathology, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhi Xie
- Medical Research Center, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jian Su
- Medical Research Center, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhi-Hong Chen
- Medical Research Center, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hong-Hong Yan
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhen Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Ben-Yuan Jiang
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Bin-Chao Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Xiao-Yan Bai
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yi-Long Wu
- Southern Medical University, Guangzhou, 510515, China.,Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.,Medical Research Center, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jin-Ji Yang
- Southern Medical University, Guangzhou, 510515, China.,Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| |
Collapse
|
193
|
Volpi CC, Gualeni AV, Pietrantonio F, Vaccher E, Carbone A, Gloghini A. Bright-field in situ hybridization detects gene alterations and viral infections useful for personalized management of cancer patients. Expert Rev Mol Diagn 2018; 18:259-277. [PMID: 29431533 DOI: 10.1080/14737159.2018.1440210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Bright-field in situ hybridization (ISH) methods detect gene alterations that may improve diagnostic precision and personalized management of cancer patients. Areas covered: This review focuses on some bright-field ISH techniques for detection of gene amplification or viral infection that have already been introduced in tumor pathology, research and diagnostic practice. Other emerging ISH methods, for the detection of translocation, mRNA and microRNA have recently been developed and need both an optimization and analytical validation. The review also deals with their clinical applications and implications on the management of cancer patients. Expert commentary: The technology of bright-field ISH applications has advanced significantly in the last decade. For example, an automated dual-color assay was developed as a clinical test for selecting cancer patients that are candidates for personalized therapy. Recently an emerging bright-field gene-protein assay has been developed. This method simultaneously detects the protein, gene and centromeric targets in the context of tissue morphology, and might be useful in assessing the HER2 status particularly in equivocal cases or samples with heterogeneous tumors. The application of bright-field ISH methods has become the gold standard for the detection of tumor-associated viral infection as diagnostic or prognostic factors.
Collapse
Affiliation(s)
- Chiara C Volpi
- a Department of Pathology and Laboratory Medicine , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| | - Ambra V Gualeni
- a Department of Pathology and Laboratory Medicine , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| | - Filippo Pietrantonio
- b Department of Medical Oncology , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| | - Emanuela Vaccher
- c Department of Medical Oncology , Centro di Riferimento Oncologico, IRCCS, National Cancer Institute , Aviano , Italy
| | - Antonino Carbone
- d Department of Pathology , Centro di Riferimento Oncologico, IRCCS, National Cancer Institute , Aviano , Italy
| | - Annunziata Gloghini
- a Department of Pathology and Laboratory Medicine , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| |
Collapse
|
194
|
Liu Q, Yu S, Zhao W, Qin S, Chu Q, Wu K. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018; 17:53. [PMID: 29455669 PMCID: PMC5817859 DOI: 10.1186/s12943-018-0793-1] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/01/2018] [Indexed: 01/29/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs)-treatments bring significant benefit for patients harboring epidermal growth factor receptor (EGFR) mutations, especially for those with lung cancer. Unfortunately, the majority of these patients ultimately develop to the acquired resistance after a period of treatment. Two central mechanisms are involved in the resistant process: EGFR secondary mutations and bypass signaling activations. In an EGFR-dependent manner, acquired mutations, such as T790 M, interferes the interaction between TKIs and the kinase domain of EGFR. While in an EGFR-independent manner, dysregulation of other receptor tyrosine kinases (RTKs) or abnormal activation of downstream compounds both have compensatory functions against the inhibition of EGFR through triggering phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK) signaling axes. Nowadays, many clinical trials aiming to overcome and prevent TKIs resistance in various cancers are ongoing or completed. EGFR-TKIs in accompany with the targeted agents for resistance-related factors afford a promising first-line strategy to further clinical application.
Collapse
Affiliation(s)
- Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
195
|
MET amplification assessed using optimized FISH reporting criteria predicts early distant metastasis in patients with non-small cell lung cancer. Oncotarget 2018; 9:12959-12970. [PMID: 29560123 PMCID: PMC5849187 DOI: 10.18632/oncotarget.24430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 01/30/2018] [Indexed: 01/02/2023] Open
Abstract
To investigate the prognostic impact of MET copy number (MET-CN) in patients with non-small cell lung cancer (NSCLC), we retrospectively reviewed clinical and pathologic data of NSCLC patients whose tumors were assessed for MET-CN using fluorescence in situ hybridization (FISH). We correlated MET-CN status with patient overall survival (OS) and optimized MET-FISH reporting criteria. The study group included 384 patients with NSCLC of which 88% were adenocarcinoma and 55.7% of patients had distant metastases. There were 170 patients with stages I-III and 214 patients with stage IV disease. Based on the MET-CN and MET/CEP7 ratio the patients were classified into 3 categories: MET-amplification (METamp): MET/CEP7 ≥ 2 or MET-CN ≥ 5; MET-CN-gain (METcng): MET-CN ≥ 4 to < 5; and MET-negative (METneg): MET-CN < 4. METamp was associated with high fatality (P=.036) and stage IV tumors (P=.038). In patients with stages I-III NSCLC, patients in the METamp category had the shortest OS (P=.015) and more often developed distant metastases within 1 year (P=.004). In patients with stage IV tumors, METamp did not further impact the OS. Patients in the METcng category had the longest OS (P=.053). Multivariate analysis confirmed METamp to be an independent high-risk factor (HR 3.26; P=.026) and predicted earlier progression to distant metastasis (HR 4.86; P=.001). In conclusion, we suggest that the MET-FISH criteria presented optimizes risk stratification by defining 3 categories of NSCLC patients. METamp is an independent risk factor predicting early distant metastasis and patients with METcng could represent a lower-risk group.
Collapse
|
196
|
Gerber DE, Socinski MA, Neal JW, Wakelee HA, Shirai K, Sequist LV, Rosovsky RP, Lilenbaum RC, Bastos BR, Huang C, Johnson ML, Hesketh PJ, Subramaniam DS, Dietrich MF, Chai F, Wang Y, Kazakin J, Schwartz B, Schiller JH, Brahmer JR, Kelly RJ. Randomized phase 2 study of tivantinib plus erlotinib versus single-agent chemotherapy in previously treated KRAS mutant advanced non-small cell lung cancer. Lung Cancer 2018; 117:44-49. [PMID: 29496255 DOI: 10.1016/j.lungcan.2018.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/16/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND KRAS mutations are identified in approximately 25% of non-small cell lung cancer (NSCLC) cases and are associated with resistance to currently available targeted therapies. The MET oncogene may be implicated in malignant progression of KRAS-mutant tumors. In a pre-specified subset analysis of KRAS mutant cancers in an earlier phase 2 study of erlotinib plus the oral MET inhibitor tivantinib, combination therapy was associated with substantial clinical benefit compared to erlotinib alone (progression-free survival [PFS] HR 0.18; P < 0.01). The current study was conducted to evaluate this combination further in KRAS mutant non-small cell lung cancer (NSCLC). MATERIALS AND METHODS Previously treated patients with advanced KRAS mutant NSCLC were randomized to receive either oral tivantinib (360 mg twice daily) plus erlotinib (150 mg daily) (ET) or single-agent chemotherapy (investigator's choice of pemetrexed, docetaxel, or gemcitabine) (C). The primary endpoint was PFS. At progression, crossover from C to ET was permitted. RESULTS Ninety-six patients were randomly assigned to ET (n = 51) or to C (n = 45). Median PFS was 1.7 months (mos) for ET and 4.3 mos for C (HR 1.19; 95% CI, 0.71-1.97; P = 0.50). There was no difference in overall survival (HR 1.20; 95% CI, 0.76-1.88; P = 0.44). There were 4 partial responses in the C arm, and none in the ET arm. Overall, adverse events occurred more frequently in the C arm, with more cytopenias, nausea, fatigue, and alopecia. Dermatologic toxicities were more common in the ET arm. CONCLUSION In previously treated patients with advanced KRAS mutant NSCLC, the combination of the MET inhibitor tivantinib and erlotinib is not superior to conventional single-agent chemotherapy.
Collapse
Affiliation(s)
- David E Gerber
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Mark A Socinski
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Joel W Neal
- Stanford University Medical Center, Palo Alto, CA, United States
| | | | - Keisuke Shirai
- Medical University of South Carolina, Charleston, SC, United States
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center, Boston, MA, United States
| | - Rachel P Rosovsky
- Massachusetts General Hospital Cancer Center, Boston, MA, United States
| | | | | | - Chao Huang
- University of Kansas Cancer Center, Westwood, KS, United States
| | | | - Paul J Hesketh
- Lahey Hospital and Medical Center, Burlington, MA, United States
| | | | - Martin F Dietrich
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Feng Chai
- ArQule, Inc., Woburn, MA, United States
| | | | | | | | - Joan H Schiller
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Julie R Brahmer
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Ronan J Kelly
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| |
Collapse
|
197
|
Thewke DP, Kou J, Fulmer ML, Xie Q. The HGF/MET Signaling and Therapeutics in Cancer. CURRENT HUMAN CELL RESEARCH AND APPLICATIONS 2018. [DOI: 10.1007/978-981-10-7296-3_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
198
|
Ai J, Chen Y, Peng X, Ji Y, Xi Y, Shen Y, Yang X, Su Y, Sun Y, Gao Y, Ma Y, Xiong B, Shen J, Ding J, Geng M. Preclinical Evaluation of SCC244 (Glumetinib), a Novel, Potent, and Highly Selective Inhibitor of c-Met in MET-dependent Cancer Models. Mol Cancer Ther 2017; 17:751-762. [DOI: 10.1158/1535-7163.mct-17-0368] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/27/2017] [Accepted: 11/30/2017] [Indexed: 11/16/2022]
|
199
|
c-Met Overexpression in Cervical Cancer, a Prognostic Factor and a Potential Molecular Therapeutic Target. Am J Clin Oncol 2017; 40:590-597. [PMID: 26083558 DOI: 10.1097/coc.0000000000000203] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE This study aimed to assess the association between pretreatment c-Met overexpression in local-regional advanced cervical cancer patients treated definitively with concurrent chemoradiation therapy (CRT) and treatment outcomes including overall survival (OS), progression-free survival (PFS), distant metastases (DM) control, and local-regional control (LC). PATIENTS AND METHODS This Institutional Review Board-approved study included cervical cancer patients treated definitively and consecutively with CRT. Evaluation of cytoplasmic immunoreactivity for c-Met was performed and scored semiquantitatively by 3 pathologists, blinded to the treatment outcomes, and incorporated both the intensity and percentage of immunoreactivity in invasive carcinoma (H score). Treatment outcomes were reviewed and reported. Outcomes were stratified by c-Met overexpression and tumor characteristics. OS, PFS, LC, and DC rates were obtained via the Kaplan-Meier method and differences between groups were evaluated by the log-rank test. Hazard ratios were obtained via Cox regression for both univariate and multivariate analyses. RESULTS The 5-year OS, PFS, LC, and DC were 57.18%, 48.07%, 72.11%, and 62.85%, respectively. Ten (35.7%) and 18 patients (64.3%) had c-Met H index >30 and<30, respectively. c-Met overexpression was significantly associated with worse 3- and 5-year OS (P=0.003), PFS (P=0.002), LC (P=0.01), and DC (P=0.0003). Patients with c-Met overexpression had a hazard ratio of 6.297, 5.782, 6.28, and 18.173 for the risks of death, disease progression, local recurrence, and DM, respectively. CONCLUSION c-Met overexpression could be a potential predictive marker and therapeutic target for local-regional advanced cervical cancer patients treated definitively with CRT.
Collapse
|
200
|
Batra U, Jain A, Sharma M, Bajaj R, Suryavanshis M. Role of crizotinib in c-mesenchymal-epidermal transition-positive nonsmall cell lung cancer patients. Indian J Cancer 2017; 54:178-181. [PMID: 29199685 DOI: 10.4103/0019-509x.219592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The increasing cases of NSCLC and improved understanding of its molecular biology has lead to identification of its varied driver mutations. cMET amplification has an important role as resistance mechanism for EGFR TKIs. Crizotinib is a drug which shows its anti-tumoral effect in cMET positive cases. Here we present a case series of three such patients who achieved were cMET amplified and showed partial response on Crizotinib.
Collapse
Affiliation(s)
- U Batra
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - A Jain
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - M Sharma
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - R Bajaj
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - M Suryavanshis
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| |
Collapse
|