351
|
Clark JW, Camidge DR, Kwak EL, Maki RG, Shapiro GI, Chen I, Tan W, Randolph S, Christensen JG, Ozeck M, Tang Y, Wilner KD, Salgia R. Dose-escalation trial of the ALK, MET & ROS1 inhibitor, crizotinib, in patients with advanced cancer. Future Oncol 2019; 16:4289-4301. [PMID: 31778074 DOI: 10.2217/fon-2019-0653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: This first-in-human, dose-finding study evaluated safety, pharmacokinetics and pharmacodynamics of crizotinib and established a recommended Phase II dose (RP2D) among patients with advanced solid malignancies. Patients & methods: Patients received oral crizotinib in a 3 + 3 dose escalation design. Results: Thirty-six patients received crizotinib (50 mg once daily-300 mg twice daily); maximum tolerated dose (and RP2D) was 250 mg twice daily. Most patients (89%) experienced ≥1 treatment-related adverse event. Three patients had grade 3 dose-limiting toxicities: alanine aminotransferase increased (n = 1) and fatigue (n = 2). Generally, an increase in soluble MET was found with increasing crizotinib concentrations. Conclusion: Crizotinib demonstrated a favorable safety profile. The observed pharmacodynamic effect on soluble MET provide evidence for targeted MET inhibition by crizotinib. Clinicaltrials. gov identifier: NCT00585195.
Collapse
Affiliation(s)
- Jeffrey W Clark
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - D Ross Camidge
- Division of Medical Oncology, University of Colorado Comprehensive Cancer Center, Aurora, CO 80045, USA
| | - Eunice L Kwak
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Robert G Maki
- Don Monti Division of Medical Oncology & Hematology, Northwell Cancer Institute & Cold Spring Harbor Laboratory, Lake Success, NY 11724, USA
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Isan Chen
- Oncology Clinical Development, Pfizer Oncology, La Jolla, CA 92121, USA
| | - Weiwei Tan
- Oncology Clinical Development, Pfizer Oncology, La Jolla, CA 92121, USA
| | - Sophia Randolph
- Oncology Clinical Development, Pfizer Oncology, La Jolla, CA 92121, USA
| | | | - Mark Ozeck
- Oncology Clinical Development, Pfizer Oncology, La Jolla, CA 92121, USA
| | - Yiyun Tang
- Oncology Clinical Development, Pfizer Oncology, La Jolla, CA 92121, USA
| | - Keith D Wilner
- Oncology Clinical Development, Pfizer Oncology, La Jolla, CA 92121, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, The City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
352
|
Guo Y, Cao R, Zhang X, Huang L, Sun L, Zhao J, Ma J, Han C. Recent Progress in Rare Oncogenic Drivers and Targeted Therapy For Non-Small Cell Lung Cancer. Onco Targets Ther 2019; 12:10343-10360. [PMID: 31819518 PMCID: PMC6886531 DOI: 10.2147/ott.s230309] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/09/2019] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is frequently associated with oncogenic driver mutations, which play an important role in carcinogenesis and cancer progression. Targeting epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase rearrangements has become standard therapy for patients with these aberrations because of the greater improvement of survival, tolerance, and quality-of-life compared to chemotherapy. Clinical trials for emerging therapies that target other less common driver genes are generating mixed results. Here, we review the literature on rare drivers in NSCLC with frequencies lower than 5% (e.g., ROS1, RET, MET, BRAF, NTRK, HER2, NRG1, FGFR1, PIK3CA, DDR2, and EGFR exon 20 insertions). In summary, targeting rare oncogenic drivers in NSCLC has achieved some success. With the development of new inhibitors that target these rare drivers, the spectrum of targeted therapy has been expanded, although acquired resistance is still an unavoidable problem.
Collapse
Affiliation(s)
- Yijia Guo
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Rui Cao
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Xiangyan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Letian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Jianzhu Zhao
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Jietao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| | - Chengbo Han
- Department of Oncology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
353
|
Sabari JK, Leonardi GC, Shu CA, Umeton R, Montecalvo J, Ni A, Chen R, Dienstag J, Mrad C, Bergagnini I, Lai WV, Offin M, Arbour KC, Plodkowski AJ, Halpenny DF, Paik PK, Li BT, Riely GJ, Kris MG, Rudin CM, Sholl LM, Nishino M, Hellmann MD, Rekhtman N, Awad MM, Drilon A. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann Oncol 2019; 29:2085-2091. [PMID: 30165371 DOI: 10.1093/annonc/mdy334] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background MET exon 14 alterations are actionable oncogenic drivers. Durable responses to MET inhibitors are observed in patients with advanced MET exon 14-altered lung cancers in prospective trials. In contrast, the activity of immunotherapy, PD-L1 expression and tumor mutational burden (TMB) of these tumors and are not well characterized. Patients and methods Patients with MET exon 14-altered lung cancers of any stage treated at two academic institutions were identified. A review of clinicopathologic and molecular features, and an analysis of response to single-agent or combination immune checkpoint inhibition were conducted. PD-L1 immunohistochemistry was carried out and TMB was calculated by estimation from targeted next-generation sequencing panels. Results We identified 147 patients with MET exon 14-altered lung cancers. PD-L1 expression of 0%, 1%-49%, and ≥50% were 37%, 22%, and 41%, respectively, in 111 evaluable tumor samples. The median TMB of MET exon 14-altered lung cancers was lower than that of unselected non-small-cell lung cancers (NSCLCs) in both independently evaluated cohorts: 3.8 versus 5.7 mutations/megabase (P < 0.001, n = 78 versus 1769, cohort A), and 7.3 versus 11.8 mutations/megabase (P < 0.001, n = 62 versus 1100, cohort B). There was no association between PD-L1 expression and TMB (Spearman's rho=0.18, P = 0.069). In response-evaluable patients (n = 24), the objective response rate was 17% (95% CI 6% to 36%) and the median progression-free survival was 1.9 months (95% CI 1.7-2.7). Responses were not enriched in tumors with PD-L1 expression ≥50% nor high TMB. Conclusion A substantial proportion of MET exon 14-altered lung cancers express PD-L1, but the median TMB is lower compared with unselected NSCLCs. Occasional responses to PD-1 blockade can be achieved, but overall clinical efficacy is modest.
Collapse
Affiliation(s)
- J K Sabari
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - G C Leonardi
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - C A Shu
- Division of Hematology Oncology, Department of Medicine, Columbia University, New York, USA
| | - R Umeton
- Department of Informatics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - J Montecalvo
- Department of Pathology, Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, New York, USA
| | - A Ni
- Department of Epidemiology and Biostatistics, Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, New York, USA
| | - R Chen
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - J Dienstag
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - C Mrad
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - I Bergagnini
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - W V Lai
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - M Offin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - K C Arbour
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - A J Plodkowski
- Department of Radiology, Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, New York, USA
| | - D F Halpenny
- Department of Radiology, Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, New York, USA
| | - P K Paik
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - B T Li
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA; Department of Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G J Riely
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - M G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - C M Rudin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - L M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - M Nishino
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Department of Radiology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M D Hellmann
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - N Rekhtman
- Department of Informatics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M M Awad
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - A Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA; Department of Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
354
|
Lenting K, van den Heuvel CNAM, van Ewijk A, ElMelik D, de Boer R, Tindall E, Wei G, Kusters B, te Dorsthorst M, ter Laan M, Huynen MA, Leenders WP. Mapping actionable pathways and mutations in brain tumours using targeted RNA next generation sequencing. Acta Neuropathol Commun 2019; 7:185. [PMID: 31747973 PMCID: PMC6865071 DOI: 10.1186/s40478-019-0826-z] [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: 09/09/2019] [Accepted: 10/02/2019] [Indexed: 01/28/2023] Open
Abstract
Many biology-based precision drugs are available that neutralize aberrant molecular pathways in cancer. Molecular heterogeneity and the lack of reliable companion diagnostic biomarkers for many drugs makes targeted treatment of cancer inaccurate for many individuals. Identifying actionable hyperactive biological pathways in individual cancers may improve this situation. To achieve this we applied a novel targeted RNA next generation sequencing (t/RNA-NGS) technique to surgically obtained glioma tissues. The test combines mutation detection with analysis of biological pathway activities that are involved in tumour behavior in many cancer types (e.g. tyrosine kinase signaling, angiogenesis signaling, immune response, metabolism), via quantitative measurement of transcript levels and splice variants of hundreds of genes. We here present proof of concept that the technique, which uses molecular inversion probes, generates a histology-independent molecular diagnosis and identifies classifiers that are strongly associated with conventional histopathology diagnoses and even with patient prognosis. The test not only confirmed known glioma-associated molecular aberrations but also identified aberrant expression levels of actionable genes and mutations that have so far been considered not to be associated with glioma, opening up the possibility of drug repurposing for individual patients. Its cost-effectiveness makes t/RNA-NGS to an attractive instrument to aid oncologists in therapy decision making.
Collapse
|
355
|
Nisa L, Francica P, Giger R, Medo M, Elicin O, Friese-Hamim M, Wilm C, Stroh C, Bojaxhiu B, Quintin A, Caversaccio MD, Dettmer MS, Buchwalder M, Brodie TM, Aebersold DM, Zimmer Y, Carey TE, Medová M. Targeting the MET Receptor Tyrosine Kinase as a Strategy for Radiosensitization in Locoregionally Advanced Head and Neck Squamous Cell Carcinoma. Mol Cancer Ther 2019; 19:614-626. [PMID: 31744898 DOI: 10.1158/1535-7163.mct-18-1274] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 08/19/2019] [Accepted: 11/14/2019] [Indexed: 11/16/2022]
Abstract
Radiotherapy (RT) along with surgery is the mainstay of treatment in head and neck squamous cell carcinoma (HNSCC). Radioresistance represents a major source of treatment failure, underlining the urgent necessity to explore and implement effective radiosensitization strategies. The MET receptor widely participates in the acquisition and maintenance of an aggressive phenotype in HNSCC and modulates the DNA damage response following ionizing radiation (IR). Here, we assessed MET expression and mutation status in primary and metastatic lesions within a cohort of patients with advanced HNSCC. Moreover, we investigated the radiosensitization potential of the MET inhibitor tepotinib in a panel of cell lines, in vitro and in vivo, as well as in ex vivo patient-derived organotypic tissue cultures (OTC). MET was highly expressed in 62.4% of primary tumors and in 53.6% of lymph node metastases (LNM), and in 6 of 9 evaluated cell lines. MET expression in primaries and LNMs was significantly associated with decreased disease control in univariate survival analyses. Tepotinib abrogated MET phosphorylation and to distinct extent MET downstream signaling. Pretreatment with tepotinib resulted in variable radiosensitization, enhanced DNA damage, cell death, and G2-M-phase arrest. Combination of tepotinib with IR led to significant radiosensitization in one of two tested in vivo models. OTCs revealed differential patterns of response toward tepotinib, irradiation, and combination of both modalities. The molecular basis of tepotinib-mediated radiosensitization was studied by a CyTOF-based single-cell mass cytometry approach, which uncovered that MET inhibition modulated PI3K activity in cells radiosensitized by tepotinib but not in the resistant ones.
Collapse
Affiliation(s)
- Lluís Nisa
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology - Head and Neck Surgery, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Paola Francica
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Roland Giger
- Department of Otorhinolaryngology - Head and Neck Surgery, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Matúš Medo
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Olgun Elicin
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Manja Friese-Hamim
- Translational Innovation Platform Oncology, Merck HealthCare KGaA, Darmstadt, Germany
| | - Claudia Wilm
- Translational Innovation Platform Oncology, Merck HealthCare KGaA, Darmstadt, Germany
| | - Christopher Stroh
- Translational Innovation Platform Oncology, Merck HealthCare KGaA, Darmstadt, Germany
| | - Beat Bojaxhiu
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Aurélie Quintin
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Marco D Caversaccio
- Department of Otorhinolaryngology - Head and Neck Surgery, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | | | - Mélanie Buchwalder
- Department of Otorhinolaryngology - Head and Neck Surgery, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Institute of Pathology, University of Bern, Bern, Switzerland
| | - Tess M Brodie
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Mass Cytometry Facility, University of Zurich, Zurich, Switzerland
| | - Daniel M Aebersold
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Yitzhak Zimmer
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Thomas E Carey
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Michaela Medová
- Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland. .,Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| |
Collapse
|
356
|
Pruis MA, Geurts-Giele WRR, von der TJH, Meijssen IC, Dinjens WNM, Aerts JGJV, Dingemans AMC, Lolkema MP, Paats MS, Dubbink HJ. Highly accurate DNA-based detection and treatment results of MET exon 14 skipping mutations in lung cancer. Lung Cancer 2019; 140:46-54. [PMID: 31862577 DOI: 10.1016/j.lungcan.2019.11.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/01/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The oncogenic MET exon 14 skipping mutation (METex14del) is described to drive 1.3 %-5.7 % of non-small-cell lung cancer (NSCLC) and multiple studies with cMET inhibitors show promising clinical responses. RNA-based analysis seems most optimal for METex14del detection, however, acquiring sufficient RNA material is often problematic. An alternative is DNA-based analysis, but commercially available DNA-based panels only detect up to 63 % of known METex14del alterations. The goal of this study is to describe an optimized DNA-based diagnostic test for METex14del in NSCLC, including clinical features and follow-up of patients treated with cMET-targeted therapy and consequent resistance mechanisms. MATERIAL AND METHODS Routinely processed diagnostic pathology non-squamous NSCLC specimens were investigated by a custom-made DNA-based targeted amplicon-based next generation sequencing (NGS) panel, which includes 4 amplicons for METex14del detection. Retrospectively, histopathological characteristics and clinical follow up were investigated for advanced non-squamous NSCLC with METex14del. RESULTS In silico analysis showed that our NGS panel is able to detect 96 % of reported METex14 alterations. METex14del was found in 2 % of patients with non-squamous NSCLC tested for therapeutic purposes. In total, from May 2015 - Sep 2018, METex14del was found in 46 patients. Thirty-six of these patients had advanced non-squamous NSCLC, they were predominantly elderly (76.5 years [53-90]), male (25/36) and (ex)-smokers (23/36). Five patients received treatment with crizotinib (Pfizer Oncology), in a named patient based program, disease control was achieved for 4/5 patients (3 partial responses, 1 stable disease) and one patient had a mixed response. Two patients developed a MET D1228N mutation during crizotinib treatment, inducing a resistance mechanism to crizotinib. CONCLUSIONS This study shows that METex14del can be reliably detected by routine DNA NGS analysis. Although a small cohort, patients responded well to targeted treatment, underlining the need for routine testing of METex14del in advanced non-squamous NSCLC to guarantee optimal personalized treatment.
Collapse
Affiliation(s)
- M A Pruis
- Department of Pulmonary Diseases, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - W R R Geurts-Giele
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Thüsen J H von der
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - I C Meijssen
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - W N M Dinjens
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - J G J V Aerts
- Department of Pulmonary Diseases, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - A M C Dingemans
- Department of Pulmonary Diseases, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Pulmonary Diseases, Maastricht UMC +, Maastricht, the Netherlands
| | - M P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - M S Paats
- Department of Pulmonary Diseases, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - H J Dubbink
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
357
|
Li Y, Ren Z, Peng Y, Li K, Wang X, Huang G, Qi S, Liu Y. Classification of glioma based on prognostic alternative splicing. BMC Med Genomics 2019; 12:165. [PMID: 31729991 PMCID: PMC6858651 DOI: 10.1186/s12920-019-0603-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Previously developed classifications of glioma have provided enormous advantages for the diagnosis and treatment of glioma. Although the role of alternative splicing (AS) in cancer, especially in glioma, has been validated, a comprehensive analysis of AS in glioma has not yet been conducted. In this study, we aimed at classifying glioma based on prognostic AS. METHODS Using the TCGA glioblastoma (GBM) and low-grade glioma (LGG) datasets, we analyzed prognostic splicing events. Consensus clustering analysis was conducted to classified glioma samples and correlation analysis was conducted to characterize regulatory network of splicing factors and splicing events. RESULTS We analyzed prognostic splicing events and proposed novel splicing classifications across pan-glioma samples (labeled pST1-7) and across GBM samples (labeled ST1-3). Distinct splicing profiles between GBM and LGG were observed, and the primary discriminator for the pan-glioma splicing classification was tumor grade. Subtype-specific splicing events were identified; one example is AS of zinc finger proteins, which is involved in glioma prognosis. Furthermore, correlation analysis of splicing factors and splicing events identified SNRPB and CELF2 as hub splicing factors that upregulated and downregulated oncogenic AS, respectively. CONCLUSION A comprehensive analysis of AS in glioma was conducted in this study, shedding new light on glioma heterogeneity and providing new insights into glioma diagnosis and treatment.
Collapse
Affiliation(s)
- Yaomin Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhonglu Ren
- College of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, 510515, Guangdong, China
| | - Yuping Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
| | - Kaishu Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiran Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China.
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Yawei Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China.
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
358
|
Liu WJ, Du Y, Wen R, Yang M, Xu J. Drug resistance to targeted therapeutic strategies in non-small cell lung cancer. Pharmacol Ther 2019; 206:107438. [PMID: 31715289 DOI: 10.1016/j.pharmthera.2019.107438] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
Rapidly developing molecular biology techniques have been employed to identify cancer driver genes in specimens from patients with non-small cell lung cancer (NSCLC). Inhibitors and antibodies that specifically target driver gene-mediated signaling pathways to suppress tumor growth and progression are expected to extend the survival time and further improve the quality of life of patients. However, the health of patients with advanced and metastatic NSCLC presents significant challenges due to treatment resistance, mediated by cancer driver gene alteration, epigenetic alteration, and tumor heterogeneity. In this review, we discuss two different resistance mechanisms in NSCLC targeted therapies, namely changes in the targeted oncogenes (on-target resistance) and changes in other related signaling pathways (off-target resistance) in tumor cells. We highlight the conventional mechanisms of drug resistance elicited by the complex heterogeneous microenvironment of NSCLC during targeted therapy, including mutations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), the receptor tyrosine kinase ROS proto-oncogene 1 (ROS1), and the serine/threonine-protein kinase BRAF (v-Raf murine sarcoma viral oncogene homolog B). We also discuss the mechanism of action of less common oncoproteins, as in-depth understanding of these molecular mechanisms is important for optimizing treatment strategies.
Collapse
Affiliation(s)
- Wen-Juan Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ru Wen
- Department of Medicine, Stanford University School of Medicine, California, USA
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, China.
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
359
|
Abstract
Papillary renal cell carcinoma (PRCC) is a subtype of renal cell carcinoma (RCC) accounting for approximately 15-20% of cases and further divided into Type 1 and Type 2. Type 1 PRCC tends to have more alterations in the MET tyrosine kinase receptor than Type 2 PRCC. Treatment for RCC patients is based on studies with minimal participation from patients with PRCC; consequently, conventional therapies tend to be less effective for RCC patients with a subtype other than ccRCC (non-ccRCC). Since MET is a known alteration in PRCC, it is potential target for directed therapy. There have been many attempts to develop MET inhibitors for use in solid tumors including PRCC. The following review will discuss the current research regarding MET-targeted therapy, MET inhibitors in clinical trials, and future directions for MET inhibitors in PRCC.
Collapse
Affiliation(s)
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
360
|
Pinsolle J, McLeer-Florin A, Giaj Levra M, de Fraipont F, Emprou C, Gobbini E, Toffart AC. Translating Systems Medicine Into Clinical Practice: Examples From Pulmonary Medicine With Genetic Disorders, Infections, Inflammations, Cancer Genesis, and Treatment Implication of Molecular Alterations in Non-small-cell Lung Cancers and Personalized Medicine. Front Med (Lausanne) 2019; 6:233. [PMID: 31737634 PMCID: PMC6828737 DOI: 10.3389/fmed.2019.00233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/03/2019] [Indexed: 12/30/2022] Open
Abstract
Non-small-cell lung cancers (NSCLC) represent 85% of all lung cancers, with adenocarcinoma as the most common subtype. Since the 2000's, the discovery of molecular alterations including epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements together with the development of specific tyrosine kinase inhibitors (TKIs) has facilitated the development of personalized medicine in the management of this disease. This review focuses on the biology of molecular alterations in NSCLC as well as the diagnostic tools and therapeutic alternatives available for each targetable alteration. Rapid and sensitive methods are essential to detect gene alterations, using tumor tissue biopsies or liquid biopsies. Massive parallel sequencing or Next Generation Sequencing (NGS) allows to simultaneously analyze numerous genes from relatively low amounts of DNA. The detection of oncogenic fusions can be conducted using fluorescence in situ hybridization, reverse-transcription polymerase chain reaction, immunohistochemistry, or NGS. EGFR mutations, ALK and ROS1 rearrangements, MET (MET proto-oncogenereceptor tyrosine kinase), BRAF (B-Raf proto-oncogen serine/threonine kinase), NTRK (neurotrophic tropomyosin receptor kinase), and RET (ret proto-oncogene) alterations are described with their respective TKIs, either already authorized or still in development. We have herein paid particular attention to the mechanisms of resistance to EGFR and ALK-TKI. As a wealth of diagnostic tools and personalized treatments are currently under development, a close collaboration between molecular biologists, pathologists, and oncologists is crucial.
Collapse
Affiliation(s)
- Julian Pinsolle
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
| | - Anne McLeer-Florin
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
- Departement of Pathological Anatomy and Cytology, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
| | - Matteo Giaj Levra
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Department of Biochemistry, Molecular Biology and Environmental Toxicology, CHU Grenoble Alpes, Grenoble, France
| | - Florence de Fraipont
- UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
- Department of Biochemistry, Molecular Biology and Environmental Toxicology, CHU Grenoble Alpes, Grenoble, France
| | - Camille Emprou
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
- Departement of Pathological Anatomy and Cytology, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
| | - Elisa Gobbini
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Cancer Research Center Lyon, Centre Léon Bérard, Lyon, France
| | - Anne-Claire Toffart
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
- UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
361
|
Duncan DJ, Vandenberghe ME, Scott MLJ, Barker C. Fast fluorescence in situ hybridisation for the enhanced detection of MET in non-small cell lung cancer. PLoS One 2019; 14:e0223926. [PMID: 31613934 PMCID: PMC6793848 DOI: 10.1371/journal.pone.0223926] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/01/2019] [Indexed: 01/29/2023] Open
Abstract
The c-Met/hepatocyte growth factor receptor pathway is frequently dysregulated in multiple cancer types, including non-small cell lung cancer (NSCLC). MET amplification has been shown to develop as a resistance mechanism to treatment in NSCLC. The identification of increased MET copy number within tumour cells is increasingly important to stratify those tumours and patients which are susceptible to treatment targetting MET kinase inhibition. Fluorescence in situ hybridisation (FISH) has been successfully employed to identify patients with abnormal MET gene copy number with numerous probes available for use. Here we report a FISH protocol that reduces probe hybridisation time in NSCLC tissue to 1 hour and compare the results with other protocols. MET gene copy number was determined in 20 NSCLC cases using 3 FISH probes: 1. Kreatech FISH, MET (7q31) SE 7 ready to use probes, hybridised using an overnight protocol; 2. Dako MET IQFISH probe with CEP7 ready to use probe, hybridised for 2 hours; 3. Kreatech MET (7q31) SE 7 XL FISH probe, prepared in SwiftFISH buffer and hybridised for 1 hour. The MET gene copy number and MET: centromere 7 gene ratio were determined for each tissue and cases categorised as having MET high or MET low status. All three FISH probes were shown to demonstrate good agreement with each other. Overall percentage agreement between probes was ≥90%. Intraclass correlation showed good agreement (ICC ≥0.80) between all three assays for MET gene copy number and MET: centromere 7 gene ratio. These FISH protocols provide evidence that rapid laboratory developed FISH assays with short turnaround time perform consistently with standard protocols, potentially enabling faster treatment decisions.
Collapse
Affiliation(s)
- David Jonathan Duncan
- Precision Medicine, R&D Oncology, AstraZeneca, Cambridge, England, United Kingdom
- * E-mail:
| | | | | | - Craig Barker
- Precision Medicine, R&D Oncology, AstraZeneca, Cambridge, England, United Kingdom
| |
Collapse
|
362
|
Baldacci S, Figeac M, Antoine M, Descarpentries C, Kherrouche Z, Jamme P, Copin MC, Tulasne D, Nanni I, Beau-Faller M, Melaabi S, Levallet G, Quoix E, Moro-Sibilot D, Friard S, Missy P, Barlesi F, Cadranel J, Cortot AB. High MET Overexpression Does Not Predict the presence of MET exon 14 Splice Mutations in NSCLC: Results From the IFCT PREDICT.amm study. J Thorac Oncol 2019; 15:120-124. [PMID: 31605799 DOI: 10.1016/j.jtho.2019.09.196] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 01/20/2023]
Abstract
INTRODUCTION MET proto-oncogene (MET) exon 14 splice site (METex14) mutations were recently described in NSCLC and has been reported to correlate with efficacy of MET tyrosine kinase inhibitors. High diversity of these alterations makes them hard to detect by DNA sequencing in clinical practice. Because METex14 mutations induce increased stabilization of the MET receptor, it is anticipated that these mutations are associated with MET overexpression. We aim to determine whether NSCLC with high MET overexpression could define a subset of patients with a high rate of METex14 mutations. METHODS From The French Cooperative Thoracic Intergroup PREDICT.amm cohort of 843 consecutive patients with a treatment-naive advanced NSCLC who were eligible for a first-line therapy, 108 NSCLC samples with high MET overexpression defined by an immunochemistry score 3+ were tested for METex14 mutations using fragment length analysis combined with optimized targeted next-generation sequencing. MET copy number analysis was also derived from the sequencing data. RESULTS METex14 mutations were detected in two patients (2.2%) who also displayed a TP53 mutation and a PIK3CA mutation, respectively. An MET gene copy number increase was observed in seven additional patients (7.7%). Next-generation sequencing analysis revealed inactivating mutations in TP53 (52.7%) and PTEN (1.1%), and oncogenic mutations in KRAS (28.6%), EGFR (7.7%), PIK3CA (4.4%), BRAF (4.4%), NRAS (2.2%), GNAS (1.1%), and IDH1 (1.1%). CONCLUSIONS The rate of METex14 mutations in NSCLC with high MET overexpression was similar to that found in unselected NSCLC. Moreover, we observed a high frequency of driver alterations in other oncogenes. Consequently these findings do not support the use of MET immunohistochemistry as a surrogate marker for METex14 mutations.
Collapse
Affiliation(s)
- Simon Baldacci
- Lille University Hospital, CHU Lille, Thoracic Oncology Department, Lille, France
| | - Martin Figeac
- University Lille, Functional and Structural Platform, CHU Lille, Lille, France
| | - Martine Antoine
- Department of Pathology, Hospital Tenon, AP-HP, Paris, France
| | - Clotilde Descarpentries
- Department of Biochemistry and Molecular Biology, Hormonology Metabolism Nutrition Oncology, CHU Lille, Lille, France
| | - Zoulika Kherrouche
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Philippe Jamme
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Marie-Christine Copin
- University Lille, CHU Lille, Institute of Pathology, UMR8161 CNRS, Institute of Biology of Lille, F-59000 Lille, France
| | - David Tulasne
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Isabelle Nanni
- Department of Molecular Oncology, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Michèle Beau-Faller
- Department of Molecular Biology, Strasbourg University Hospital, Strasbourg, France
| | - Samia Melaabi
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | | | - Elisabeth Quoix
- Department of Pneumology, Nouvel Hôpital Civil, University Hospital, Strasbourg, France
| | - Denis Moro-Sibilot
- Department of Pneumology, Grenoble University Hospital, Grenoble, France
| | - Sylvie Friard
- Pneumology Department, Foch Hospital, Suresnes, France
| | - Pascale Missy
- Clinical Research Unit, French Cooperative Thoracic Intergroup (IFCT), Paris, France
| | - Fabrice Barlesi
- Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Multidisciplinary Oncology & Therapeutic Innovations Department, Marseille, France
| | - Jacques Cadranel
- Chest Department-Thoracic Oncology Expert Center, AP-HP, Groupe Hospitalier HUEP, Hopital Tenon, Paris, France, and Sorbonne University, Paris, France
| | - Alexis B Cortot
- Lille University Hospital, CHU Lille, Thoracic Oncology Department, Lille, France.
| |
Collapse
|
363
|
Comparative analysis of expression of mutant and wild-type alleles is essential for reliable PCR-based detection of MET exon 14 skipping. Biochimie 2019; 165:267-274. [DOI: 10.1016/j.biochi.2019.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/26/2019] [Indexed: 01/05/2023]
|
364
|
Hong W, Zhang W, Guan R, Liang Y, Hu S, Ji Y, Liu M, Lu H, Yu M, Ma L. Genome-wide profiling of prognosis-related alternative splicing signatures in sarcoma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:557. [PMID: 31807538 PMCID: PMC6861818 DOI: 10.21037/atm.2019.09.65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Sarcomas (SARCs) are rare malignant tumors with poor prognosis. Increasing evidence has suggested that aberrant alternative splicing (AS) is strongly associated with tumor initiation and progression. We considered whether survival-related AS events might serve as prognosis predictors and underlying targeted molecules in SARC treatment. METHODS RNA-Seq data of the SARC cohort were downloaded from The Cancer Genome Atlas (TCGA) database. Survival-related AS events were selected by univariate and multivariate Cox regression analyses. Metascape was used for constructing a gene interaction network and performing functional enrichment analysis. Then, prognosis predictors were established based on statistically significant survival-related AS events and evaluated by receiver operator characteristic (ROC) curve analysis. Finally, the potential regulatory network was analyzed via Pearson's correlation between survival-related AS events and splicing factors (SFs). RESULTS A total of 3,610 AS events and 2,291 genes were found to be prognosis-related in 261 SARC samples. The focal adhesion pathway was identified as the most critical molecular mechanism corresponding to poor prognosis. Notably, several prognosis predictors based on survival-related AS events showed excellent performance in prognosis prediction. The area under the curve of the ROC of the risk score was 0.85 in the integrated predictor. The splicing network proved complicated regulation between prognosis-related SFs and AS events. Also, driver gene mutations were significantly associated with AS in SARC patients. CONCLUSIONS Survival-related AS events may become ideal indictors for the prognosis prediction of SARCs. Corresponding splicing regulatory mechanisms are worth further exploration.
Collapse
Affiliation(s)
- Weifeng Hong
- Department of Medical Imaging, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Weicong Zhang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Renguo Guan
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, China
| | - Yuying Liang
- Department of Medical Imaging, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Shixiong Hu
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, China
| | - Yayun Ji
- Department of Medical Imaging, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Mouyuan Liu
- Department of Medical Imaging, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Hai Lu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Min Yu
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, China
| | - Liheng Ma
- Department of Medical Imaging, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| |
Collapse
|
365
|
Keppens C, Tack V, Dufraing K, Rouleau E, Ligtenberg MJL, Schuuring E, Dequeker EMC. Variation in nomenclature of somatic variants for selection of oncological therapies: Can we reach a consensus soon? Hum Mutat 2019; 41:7-16. [PMID: 31553104 PMCID: PMC6973115 DOI: 10.1002/humu.23926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/05/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022]
Abstract
A standardized nomenclature for reporting oncology biomarker variants is key to avoid misinterpretation of results and unambiguous registration in clinical databases. External quality assessment (EQA) schemes have revealed a need for more consistent nomenclature use in clinical genetics. We evaluated the propensity of EQA for improvement of compliance with Human Genome Variation Society (HGVS) recommendations for reporting of predictive somatic variants in lung and colorectal cancer. Variant entries between 2012 and 2018 were collected from written reports and electronic results sheets. In total, 4,053 variants were assessed, of which 12.1% complied with HGVS recommendations. Compliance improved over time from 2.1% (2012) to 22.3% (2018), especially when laboratories participated in multiple EQA schemes. Compliance was better for next-generation sequencing (20.9%) compared with targeted techniques (9.8%). In the 1792 reports, HGVS recommendations for reference sequences were met for 31.9% of reports, for 36.0% of noncommercial, and 26.5% of commercial test methods. Compliance improved from 16.7% (2012) to 33.1% (2018), and after repeated EQA participation. EQA participation improves compliance with HGVS recommendations. The residual percentage of errors in the most recent schemes suggests that laboratories, companies, and EQA providers need to collaborate for additional improvement of harmonization in clinical test reporting.
Collapse
Affiliation(s)
- Cleo Keppens
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Véronique Tack
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Kelly Dufraing
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| | - Etienne Rouleau
- Gustave Roussy, Service de Génétique/Pathologie Moléculaire, Villejuif Cedex, France
| | - Marjolijn J L Ligtenberg
- Department of Pathology and Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ed Schuuring
- University Medical Center Groningen, Department of Pathology, University of Groningen, Groningen, The Netherlands
| | - Elisabeth M C Dequeker
- Department of Public Health and Primary Care, Biomedical Quality Assurance Research Unit, University of Leuven, Leuven, Belgium
| |
Collapse
|
366
|
Rotow JK, Gui P, Wu W, Raymond VM, Lanman RB, Kaye FJ, Peled N, Fece de la Cruz F, Nadres B, Corcoran RB, Yeh I, Bastian BC, Starostik P, Newsom K, Olivas VR, Wolff AM, Fraser JS, Collisson EA, McCoach CE, Camidge DR, Pacheco J, Bazhenova L, Li T, Bivona TG, Blakely CM. Co-occurring Alterations in the RAS-MAPK Pathway Limit Response to MET Inhibitor Treatment in MET Exon 14 Skipping Mutation-Positive Lung Cancer. Clin Cancer Res 2019; 26:439-449. [PMID: 31548343 DOI: 10.1158/1078-0432.ccr-19-1667] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/19/2019] [Accepted: 09/10/2019] [Indexed: 01/07/2023]
Abstract
PURPOSE Although patients with advanced-stage non-small cell lung cancers (NSCLC) harboring MET exon 14 skipping mutations (METex14) often benefit from MET tyrosine kinase inhibitor (TKI) treatment, clinical benefit is limited by primary and acquired drug resistance. The molecular basis for this resistance remains incompletely understood. EXPERIMENTAL DESIGN Targeted sequencing analysis was performed on cell-free circulating tumor DNA obtained from 289 patients with advanced-stage METex14-mutated NSCLC. RESULTS Prominent co-occurring RAS-MAPK pathway gene alterations (e.g., in KRAS, NF1) were detected in NSCLCs with METex14 skipping alterations as compared with EGFR-mutated NSCLCs. There was an association between decreased MET TKI treatment response and RAS-MAPK pathway co-occurring alterations. In a preclinical model expressing a canonical METex14 mutation, KRAS overexpression or NF1 downregulation hyperactivated MAPK signaling to promote MET TKI resistance. This resistance was overcome by cotreatment with crizotinib and the MEK inhibitor trametinib. CONCLUSIONS Our study provides a genomic landscape of co-occurring alterations in advanced-stage METex14-mutated NSCLC and suggests a potential combination therapy strategy targeting MAPK pathway signaling to enhance clinical outcomes.
Collapse
Affiliation(s)
- Julia K Rotow
- Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Philippe Gui
- Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Wei Wu
- Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | | | | | - Frederic J Kaye
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Nir Peled
- Soroka Medical Center, Ben-Gurion University, Beer-Sheva, Israel
| | - Ferran Fece de la Cruz
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Brandon Nadres
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ryan B Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Iwei Yeh
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Departments of Dermatology and Pathology, and Clinical Cancer Genomics Laboratory, University of California, San Francisco, California
| | - Boris C Bastian
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Departments of Dermatology and Pathology, and Clinical Cancer Genomics Laboratory, University of California, San Francisco, California
| | - Petr Starostik
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Kimberly Newsom
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Victor R Olivas
- Department of Medicine, University of California, San Francisco, California
| | - Alexander M Wolff
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| | - Eric A Collisson
- Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Caroline E McCoach
- Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | | | | | | | - Tianhong Li
- Department of Internal Medicine, University of California, Davis, California
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| |
Collapse
|
367
|
Heydt C, Becher AK, Wagener-Ryczek S, Ball M, Schultheis AM, Schallenberg S, Rüsseler V, Büttner R, Merkelbach-Bruse S. Comparison of in situ and extraction-based methods for the detection of MET amplifications in solid tumors. Comput Struct Biotechnol J 2019; 17:1339-1347. [PMID: 31762957 PMCID: PMC6861603 DOI: 10.1016/j.csbj.2019.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 01/22/2023] Open
Abstract
In EGFR-treatment naive NSCLC patients, high-level MET amplification is detected in approximately 2-3% and is considered as adverse prognostic factor. Currently, clinical trials with two different inhibitors, capmatinib and tepotinib, are under way both defining different inclusion criteria regarding MET amplification from proven amplification only to defining an exact MET copy number. Here, 45 patient samples, including 10 samples without MET amplification, 5 samples showing a low-level MET amplification, 10 samples with an intermediate-level MET amplification, 10 samples having a high-level MET amplification by a MET/CEN7 ratio ≥2.0 and 10 samples showing a high-level MET amplification with GCN ≥6, were evaluated by MET FISH, MET IHC, a ddPCR copy number assay, a NanoString nCounter copy number assay and an amplicon-based parallel sequencing. The MET IHC had the best concordance with MET FISH followed by the NanoString copy number assay, the ddPCR copy number assay and the custom amplicon-based parallel sequencing assays. The concordance was higher in the high-level amplified cohorts than in the low- and intermediate-level amplified cohorts. In summary, currently extraction-based methods cannot replace the MET FISH for the detection of low-level, intermediate-level and high-level MET amplifications, as the number of false negative results is very high. Only for the detection of high-level amplified samples with a gene copy number ≥6 extraction-based methods are a reliable alternative.
Collapse
Affiliation(s)
- Carina Heydt
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Ann-Kathrin Becher
- Institute of Pathology, University Hospital Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Svenja Wagener-Ryczek
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Markus Ball
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Anne M. Schultheis
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Simon Schallenberg
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Vanessa Rüsseler
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
368
|
Jiang W, Yang N, Zhang Y. Novel MET Exon 14 Skipping Treatment-Naïve Lung Adenocarcinoma Presented Primary Resistance to Crizotinib. J Thorac Oncol 2019; 13:e124-e126. [PMID: 29935852 DOI: 10.1016/j.jtho.2018.02.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Wenjuan Jiang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, China.
| |
Collapse
|
369
|
He AR, Cohen RB, Denlinger CS, Sama A, Birnbaum A, Hwang J, Sato T, Lewis N, Mynderse M, Niland M, Giles J, Wallin J, Moser B, Zhang W, Walgren R, Plimack ER. First-in-Human Phase I Study of Merestinib, an Oral Multikinase Inhibitor, in Patients with Advanced Cancer. Oncologist 2019; 24:e930-e942. [PMID: 30833489 PMCID: PMC6738318 DOI: 10.1634/theoncologist.2018-0411] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The purpose of this nonrandomized, open-label, phase I study (NCT01285037) was to evaluate the safety and tolerability of merestinib, an oral antiproliferative and antiangiogenic kinase inhibitor, and to determine a recommended phase II dose and schedule for patients with advanced cancer. MATERIALS AND METHODS This was a multicenter, nonrandomized, open-label, phase I study of oral merestinib consisting of six parts: dose escalation (part A), followed by a four-cohort dose-confirmation study (part B) and subsequently a four-part dose expansion and combination safety testing of merestinib with standard doses of cetuximab (part C), cisplatin (part D), gemcitabine and cisplatin (part E), and ramucirumab (part F) in patients with specific types of advanced cancers. Safety, tolerability, antitumor activity, and pharmacokinetics were evaluated in all cohorts. RESULTS The dose escalation, confirmation, and expansion results support the dosing of merestinib at 120 mg once daily, based on acceptable exposure and safety at this dose. One complete response was observed in a patient with cholangiocarcinoma, and three patients with cholangiocarcinoma achieved a partial response. Overall, 60 (32%) of the 186 patients enrolled in the study had a best response of stable disease. CONCLUSION This study demonstrates that merestinib has a tolerable safety profile and potential anticancer activity and warrants further clinical investigation. IMPLICATIONS FOR PRACTICE Merestinib treatment in patients with advanced cancer demonstrated an acceptable safety profile and potential antitumor activity, supporting its future development in specific disease populations as a monotherapy and/or in combination with other therapies.
Collapse
Affiliation(s)
- Aiwu Ruth He
- Department of Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Roger B Cohen
- Perelman School of Medicine at the University of Pennsylvania, Abramson Cancer Center, Philadelphia, Pennsylvania, USA
| | - Crystal S Denlinger
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania, USA
| | - Ashwin Sama
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ariel Birnbaum
- Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Jimmy Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, USA
| | - Takami Sato
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nancy Lewis
- Translational Clinical Oncology, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | | | | | | | - Brian Moser
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Wei Zhang
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania, USA
| |
Collapse
|
370
|
Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer 2019; 19:495-509. [PMID: 31406302 PMCID: PMC7043073 DOI: 10.1038/s41568-019-0179-8] [Citation(s) in RCA: 655] [Impact Index Per Article: 109.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 02/07/2023]
Abstract
The impressive clinical activity of small-molecule receptor tyrosine kinase inhibitors for oncogene-addicted subgroups of non-small-cell lung cancer (for example, those driven by activating mutations in the gene encoding epidermal growth factor receptor (EGFR) or rearrangements in the genes encoding the receptor tyrosine kinases anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 (ROS1) and rearranged during transfection (RET)) has established an oncogene-centric molecular classification paradigm in this disease. However, recent studies have revealed considerable phenotypic diversity downstream of tumour-initiating oncogenes. Co-occurring genomic alterations, particularly in tumour suppressor genes such as TP53 and LKB1 (also known as STK11), have emerged as core determinants of the molecular and clinical heterogeneity of oncogene-driven lung cancer subgroups through their effects on both tumour cell-intrinsic and non-cell-autonomous cancer hallmarks. In this Review, we discuss the impact of co-mutations on the pathogenesis, biology, microenvironmental interactions and therapeutic vulnerabilities of non-small-cell lung cancer and assess the challenges and opportunities that co-mutations present for personalized anticancer therapy, as well as the expanding field of precision immunotherapy.
Collapse
Affiliation(s)
- Ferdinandos Skoulidis
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
371
|
Landi L, Chiari R, Tiseo M, D'Incà F, Dazzi C, Chella A, Delmonte A, Bonanno L, Giannarelli D, Cortinovis DL, de Marinis F, Borra G, Morabito A, Gridelli C, Galetta D, Barbieri F, Grossi F, Capelletto E, Minuti G, Mazzoni F, Verusio C, Bria E, Alì G, Bruno R, Proietti A, Fontanini G, Crinò L, Cappuzzo F. Crizotinib in MET-Deregulated or ROS1-Rearranged Pretreated Non–Small Cell Lung Cancer (METROS): A Phase II, Prospective, Multicenter, Two-Arms Trial. Clin Cancer Res 2019; 25:7312-7319. [DOI: 10.1158/1078-0432.ccr-19-0994] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/13/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
|
372
|
Hewelt B, Li H, Jolly MK, Kulkarni P, Mambetsariev I, Salgia R. The DNA walk and its demonstration of deterministic chaos-relevance to genomic alterations in lung cancer. Bioinformatics 2019; 35:2738-2748. [PMID: 30615123 PMCID: PMC6691335 DOI: 10.1093/bioinformatics/bty1021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/05/2018] [Accepted: 01/04/2019] [Indexed: 12/25/2022] Open
Abstract
MOTIVATION Advancements in cancer genetics have facilitated the development of therapies with actionable mutations. Although mutated genes have been studied extensively, their chaotic behavior has not been appreciated. Thus, in contrast to naïve DNA, mutated DNA sequences can display characteristics of unpredictability and sensitivity to the initial conditions that may be dictated by the environment, expression patterns and presence of other genomic alterations. Employing a DNA walk as a form of 2D analysis of the nucleotide sequence, we demonstrate that chaotic behavior in the sequence of a mutated gene can be predicted. RESULTS Using fractal analysis for these DNA walks, we have determined the complexity and nucleotide variance of commonly observed mutated genes in non-small cell lung cancer, and their wild-type counterparts. DNA walks for wild-type genes demonstrate varying levels of chaos, with BRAF, NTRK1 and MET exhibiting greater levels of chaos than KRAS, paxillin and EGFR. Analyzing changes in chaotic properties, such as changes in periodicity and linearity, reveal that while deletion mutations indicate a notable disruption in fractal 'self-similarity', fusion mutations demonstrate bifurcations between the two genes. Our results suggest that the fractals generated by DNA walks can yield important insights into potential consequences of these mutated genes. AVAILABILITY AND IMPLEMENTATION Introduction to Turtle graphics in Python is an open source article on learning to develop a script for Turtle graphics in Python, freely available on the web at https://docs.python.org/2/library/turtle.html. cDNA sequences were obtained through NCBI RefSeq database, an open source database that contains information on a large array of genes, such as their nucleotide and amino acid sequences, freely available at https://www.ncbi.nlm.nih.gov/refseq/. FracLac plugin for Fractal analysis in ImageJ is an open source plugin for the ImageJ program to perform fractal analysis, free to download at https://imagej.nih.gov/ij/plugins/fraclac/FLHelp/Introduction.html. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Blake Hewelt
- Department of Medical Oncology and Therapeutics Research
| | - Haiqing Li
- Department of Computational & Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | | | | | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research
| |
Collapse
|
373
|
Cao J, Chen L, Li H, Chen H, Yao J, Mu S, Liu W, Zhang P, Cheng Y, Liu B, Hu Z, Chen D, Kang H, Hu J, Wang A, Wang W, Yao M, Chrin G, Wang X, Zhao W, Li L, Xu L, Guo W, Jia J, Chen J, Wang K, Li G, Shi W. An Accurate and Comprehensive Clinical Sequencing Assay for Cancer Targeted and Immunotherapies. Oncologist 2019; 24:e1294-e1302. [PMID: 31409745 DOI: 10.1634/theoncologist.2019-0236] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/25/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Incorporation of next-generation sequencing (NGS) technology into clinical utility in targeted and immunotherapies requires stringent validation, including the assessment of tumor mutational burden (TMB) and microsatellite instability (MSI) status by NGS as important biomarkers for response to immune checkpoint inhibitors. MATERIALS AND METHODS We designed an NGS assay, Cancer Sequencing YS panel (CSYS), and applied algorithms to detect five classes of genomic alterations and two genomic features of TMB and MSI. RESULTS By stringent validation, CSYS exhibited high sensitivity and predictive positive value of 99.7% and 99.9%, respectively, for single nucleotide variation; 100% and 99.9%, respectively, for short insertion and deletion (indel); and 95.5% and 100%, respectively, for copy number alteration (CNA). Moreover, CSYS achieved 100% specificity for both long indel (50-3,000 bp insertion and deletion) and gene rearrangement. Overall, we used 33 cell lines and 208 clinical samples to validate CSYS's NGS performance, and genomic alterations in clinical samples were also confirmed by fluorescence in situ hybridization, immunohistochemistry, and polymerase chain reaction (PCR). Importantly, the landscape of TMB across different cancers of Chinese patients (n = 3,309) was studied. TMB by CSYS exhibited a high correlation (Pearson correlation coefficient r = 0.98) with TMB by whole exome sequencing (WES). MSI measurement showed 98% accuracy and was confirmed by PCR. Application of CSYS in a clinical setting showed an unexpectedly high occurrence of long indel (6.3%) in a cohort of tumors from Chinese patients with cancer (n = 3,309), including TP53, RB1, FLT3, BRCA2, and other cancer driver genes with clinical impact. CONCLUSION CSYS proves to be clinically applicable and useful in disclosing genomic alterations relevant to cancer target therapies and revealing biomarkers for immune checkpoint inhibitors. IMPLICATIONS FOR PRACTICE The study describes a specially designed sequencing panel assay to detect genomic alterations and features of 450 cancer genes, including its overall workflow and rigorous clinical and analytical validations. The distribution of pan-cancer tumor mutational burden, microsatellite instability, gene rearrangement, and long insertion and deletion mutations was assessed for the first time by this assay in a broad array of Chinese patients with cancer. The Cancer Sequencing YS panel and its validation study could serve as a blueprint for developing next-generation sequencing-based assays, particularly for the purpose of clinical application.
Collapse
Affiliation(s)
- Jingyu Cao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Lijuan Chen
- OrigiMed, Shanghai, People's Republic of China
| | - Heng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, People's Republic of China
| | - Hui Chen
- OrigiMed, Shanghai, People's Republic of China
| | - Jicheng Yao
- OrigiMed, Shanghai, People's Republic of China
| | - Shuo Mu
- OrigiMed, Shanghai, People's Republic of China
| | - Wenjin Liu
- OrigiMed, Shanghai, People's Republic of China
| | - Peng Zhang
- OrigiMed, Shanghai, People's Republic of China
| | - Yuwei Cheng
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
| | - Binbin Liu
- OrigiMed, Shanghai, People's Republic of China
| | | | | | - Hui Kang
- OrigiMed, Shanghai, People's Republic of China
| | - Jinwei Hu
- OrigiMed, Shanghai, People's Republic of China
| | - Aodi Wang
- OrigiMed, Shanghai, People's Republic of China
| | | | - Ming Yao
- OrigiMed, Shanghai, People's Republic of China
| | | | - Xiaoting Wang
- Department of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, People's Republic of China
| | - Wei Zhao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Lei Li
- Department of Hepatobiliary Surgery, Shandong Tumor Hospital, Jinan, People's Republic of China
| | - Luping Xu
- Department of General Surgery, The First Affiliated Hospital, Jiaxing College of Medicine, Jiangxi, People's Republic of China
| | - Weixin Guo
- Department of Chemotherapy, Meizhou People's Hospital, Meizhou, People's Republic of China
| | - Jun Jia
- Department of Oncology, Dongguan People's Hospital, Dongguan, People's Republic of China
| | - Jianhua Chen
- Department of Medical Oncology-Chest, Hunan Cancer Hospital, Changsha, People's Republic of China
| | - Kai Wang
- OrigiMed, Shanghai, People's Republic of China
- Zhejiang University International Hospital, Hangzhou, People's Republic of China
| | - Gaofeng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, People's Republic of China
| | - Weiwei Shi
- OrigiMed, Shanghai, People's Republic of China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
374
|
Chu LP, Franck D, Parachoniak CA, Gregg JP, Moore MG, Farwell DG, Rao S, Heilmann AM, Erlich RL, Ross JS, Miller VA, Ali S, Riess JW. MET Genomic Alterations in Head and Neck Squamous Cell Carcinoma (HNSCC): Rapid Response to Crizotinib in a Patient with HNSCC with a Novel MET R1004G Mutation. Oncologist 2019; 24:1305-1308. [PMID: 31391294 DOI: 10.1634/theoncologist.2019-0230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022] Open
Abstract
Identification of effective targeted therapies for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) remains an unmet medical need. A patient with platinum-refractory recurrent oral cavity HNSCC underwent comprehensive genomic profiling (CGP) that identified an activating MET mutation (R1004). The patient was treated with the oral MET tyrosine kinase inhibitor crizotinib with rapid response to treatment.Based on this index case, we determined the frequency of MET alterations in 1,637 HNSCC samples, which had been analyzed with hybrid capture-based CGP performed in the routine course of clinical care. The specimens were sequenced to a median depth of >500× for all coding exons from 182 (version 1, n = 24), 236 (version 2, n = 326), or 315 (version 3, n = 1,287) cancer-related genes, plus select introns from 14 (version 1), 19 (version 2), or 28 (version 3) genes frequently rearranged in cancer. We identified 13 HNSCC cases (0.79%) with MET alterations (4 point mutation events and 9 focal amplification events). MET-mutant or amplified tumors represent a small but potentially actionable molecular subset of HNSCC. KEY POINTS: This case report is believed to be the first reported pan-cancer case of a patient harboring a MET mutation at R1004 demonstrating a clinical response to crizotinib, in addition to the first documented case of head and neck squamous cell carcinoma (HNSCC) with any MET alteration responding to crizotinib.The positive response to MET inhibition in this patient highlights the significance of comprehensive genomic profiling in advanced metastatic HNSCC to identify actionable targetable molecular alterations as current treatment options are limited.
Collapse
Affiliation(s)
- Lisa Pei Chu
- Department of Internal Medicine, Division of Hematology/Oncology, UC Davis School of Medicine, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Debra Franck
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Jeffrey P Gregg
- Department of Pathology, UC Davis School of Medicine, Sacramento, California, USA
| | - Michael G Moore
- Department of Otolaryngology, UC Davis School of Medicine, Sacramento, California, USA
| | - D Gregory Farwell
- Department of Otolaryngology, UC Davis School of Medicine, Sacramento, California, USA
| | - Shyam Rao
- Department of Radiation Oncology, UC Davis School of Medicine, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | | | | | | | | | - Siraj Ali
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Jonathan W Riess
- Department of Internal Medicine, Division of Hematology/Oncology, UC Davis School of Medicine, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| |
Collapse
|
375
|
Pan Y, Zhang Y, Ye T, Zhao Y, Gao Z, Yuan H, Zheng D, Zheng S, Li H, Li Y, Jin Y, Sun Y, Chen H. Detection of Novel NRG1, EGFR, and MET Fusions in Lung Adenocarcinomas in the Chinese Population. J Thorac Oncol 2019; 14:2003-2008. [PMID: 31382039 DOI: 10.1016/j.jtho.2019.07.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Multiple oncogene fusions beyond ALK receptor tyrosine kinase (ALK), RET, and ROS1 fusion has been described in lung cancer, especially in lung adenocarcinomas without common oncogenic mutations. Molecular inhibitors have been developed and proved effective for patients whose tumors harbor these novel alterations. METHODS A consecutive series of surgically resected lung adenocarcinomas were collected and profiled using an enrichment strategy to detect nine common oncogenic driver mutations and fusions concerning EGFR, KRAS, HER2, BRAF, MET, ALK, RET, ROS1, and FGFR. Driver-negative cases were further analyzed by a comprehensive RNA-based next-generation sequencing (NGS) fusion assay for novel fusions. RESULTS In total, we profiled 1681 lung adenocarcinomas, among which 255 cases were common driver-negative. One hundred seventy-seven cases had sufficient tissue for NGS fusions screening, which identified eight novel fusions. NRG1 fusions occurred in 0.36% of all lung adenocarcinoma cases (6 of 1681 cases), including 4 CD74-NRG1-positive cases, 1 RBPMS-NRG1-positive case, and 1 novel ITGB1-NRG1-positive case. Furthermore, another 2 novel fusions were also detected, including 1 EGFR-SHC1 fusion and 1 CD47-MET fusion, both of which were in-frame and retained the functional domain of the corresponding kinases. No fusion event was detected for NTRK, KRAS, BRAF or HER2 genes in this cohort. Detailed clinicopathologic data showed that invasive mucous adenocarcinoma (three of eight cases) and acinar-predominant adenocarcinoma (three of eight cases) were the most prevalent pathologic subtypes among novel fusions. CONCLUSIONS Fusions affecting NRG1, EGFR, and MET were detected in 0.48% of unselected lung adenocarcinomas, and NRG1 fusions ranked the most prevalent fusions in common driver-negative lung adenocarcinomas from Chinese population. RNA-based NGS fusion assay was an optional method for screening actionable fusions in common driver-negative cases.
Collapse
Affiliation(s)
- Yunjian Pan
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yang Zhang
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ting Ye
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yue Zhao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhendong Gao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hui Yuan
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Difan Zheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shanbo Zheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hang Li
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuan Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yan Jin
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yihua Sun
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China; Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
376
|
Vansteenkiste JF, Van De Kerkhove C, Wauters E, Van Mol P. Capmatinib for the treatment of non-small cell lung cancer. Expert Rev Anticancer Ther 2019; 19:659-671. [PMID: 31368815 DOI: 10.1080/14737140.2019.1643239] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction: Activation of the MET pathway through MET amplifications or mutations is present in 3-4% of stage IV non-squamous non-small cell lung cancers (NSCLC). High MET amplifications and exon 14 skipping mutations are associated with poor prognosis: new treatments are needed for these patients. Capmatinib is a highly selective, potent small-molecule MET inhibitor with antitumor activity in NSCLC in vitro and in vivo. Areas covered: This article provides an overview of the capmatinib clinical development program in NSCLC, both as monotherapy in NSCLC with a dysregulated MET pathway, and in combination with epidermal growth factor receptor (EGFR) inhibitor therapy in EGFR-mutant NSCLC with MET-based acquired resistance to previous EGFR inhibition. Expert opinion: In the GEOMETRY Mono-1 study, treatment with capmatinib resulted in high response rates in stage IV NSCLC with MET exon 14 skipping mutations, particularly in first line, supporting testing for this biomarker at the time of diagnosis. Durable responses have been reported and results in MET-amplified NSCLC are eagerly anticipated. In EGFR-mutant NSCLC, notable responses have been observed in combination with an EGFR-tyrosine kinase inhibitor (TKI) in case of acquired resistance to EGFR-TKIs based on high MET amplification.
Collapse
Affiliation(s)
| | | | - Els Wauters
- Respiratory Oncology Unit (Respiratory Diseases), University Hospital KU Leuven , Leuven , Belgium
| | - Pierre Van Mol
- Respiratory Oncology Unit (Respiratory Diseases), University Hospital KU Leuven , Leuven , Belgium
| |
Collapse
|
377
|
Lung J, Hung MS, Lin YC, Lee KF, Jiang YY, Huang SL, Fang YH, Lu MS, Lin CK, Yang TM, Lin PY, Hsieh MJ, Tsai YH. MET exon 14 skipping mutations and gene amplification in a Taiwanese lung cancer population. PLoS One 2019; 14:e0220670. [PMID: 31369639 PMCID: PMC6675391 DOI: 10.1371/journal.pone.0220670] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/20/2019] [Indexed: 12/21/2022] Open
Abstract
Somatic mutations of MET gene are emerging as important driver mutations for lung cancers. To identify the common clinicopathological features of MET exon 14 skipping mutations and amplification and clarify whether the two MET gene alterations cause protein overexpression were investigated using 196 lung cancer samples of Taiwan through real time-qPCR/sequencing, fluorescence in situ hybridization, and immunohistochemistry. The two MET gene alterations are both present in low frequency, ~1%, in the studied lung cancer population of Taiwan. MET exon 14 skipping mutations were identified from two early-stage patients, who were both relatively advanced in age, and did not carry other driver mutations. One was an adenocarcinoma and the other was a rare carcinosarcoma. Three gene amplifications cases were identified. Neither of the two MET gene alterations would lead to protein overexpression; hence, direct detection in nucleic acid level would be a preferred and straightforward solution for the identification of skipping mutations. The presence of MET exon 14 mutations in minor histological types of lung cancers urge to extend screening scope of this mutation in lung cancer and treatment response evaluation in clinical trials. These would be important next steps for the success of MET target therapy in clinical practice.
Collapse
Affiliation(s)
- Jrhau Lung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Ming-Szu Hung
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Yu-Ching Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Kam-Fai Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Yuan Yuan Jiang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Shao-Lan Huang
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Yu-Hung Fang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Ming-Shian Lu
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Chin-Kuo Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Tsung-Ming Yang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
| | - Paul Yann Lin
- Department of Anatomic Pathology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Meng-Jer Hsieh
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Respiratory Care, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying Huang Tsai
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch, Taiwan
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taiwan
- * E-mail:
| |
Collapse
|
378
|
Hong S, Liu D, Luo S, Fang W, Zhan J, Fu S, Zhang Y, Wu X, Zhou H, Chen X, Chen G, Zhang Z, Zheng Q, Li X, Chen J, Liu X, Lei M, Ye C, Wang J, Yang H, Xu X, Zhu S, Yang Y, Zhao Y, Zhou N, Zhao H, Huang Y, Zhang L, Wu K, Zhang L. The genomic landscape of Epstein-Barr virus-associated pulmonary lymphoepithelioma-like carcinoma. Nat Commun 2019; 10:3108. [PMID: 31311932 PMCID: PMC6635366 DOI: 10.1038/s41467-019-10902-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Pulmonary lymphoepithelioma-like carcinoma (LELC) is a rare and distinct subtype of primary lung cancer characterized by Epstein-Barr virus (EBV) infection. Herein, we reported the mutational landscape of pulmonary LELC using whole-exome sequencing, targeted deep sequencing and single-nucleotide polymorphism arrays. We identify a low degree of somatic mutation but widespread existence of copy number variations. We reveal predominant signature 2 mutations and frequent loss of type I interferon genes that are involved in the host-virus counteraction. Integrated analysis shows enrichment of genetic lesions affecting several critical pathways, including NF-κB, JAK/STAT, and cell cycle. Notably, multi-dimensional comparison unveils that pulmonary LELC resemble NPC but are clearly different from other lung cancers, natural killer/T-cell lymphoma or EBV-related gastric cancer in terms of genetic features. In all, our study illustrates a distinct genomic landscape of pulmonary LELC and provides a road map to facilitate genome-guided personalized treatment. The rare lung cancer subtype pulmonary lymphoepithelioma-like carcinoma is linked to Epstein-Barr virus infection. Here, the authors provide a mutational landscape for this cancer, showing a low burden of somatic mutations and high prevalence of copy number variations.
Collapse
Affiliation(s)
- Shaodong Hong
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Dongbing Liu
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Shuzhen Luo
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Jianhua Zhan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Sha Fu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, 510120, Guangzhou, China
| | - Yaxiong Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Xuan Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Huaqiang Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Xi Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Gang Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Zhonghan Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Qiufan Zheng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Xiaobo Li
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China.,BGI Education Center, University of Chinese Academy of Sciences, 518083, Shenzhen, China
| | - Jinghao Chen
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China.,BGI Education Center, University of Chinese Academy of Sciences, 518083, Shenzhen, China
| | | | - Mengyue Lei
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Chen Ye
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Jian Wang
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Huanming Yang
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Xun Xu
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Shida Zhu
- BGI-Shenzhen, 518083, Shenzhen, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Ningning Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Hongyun Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Lanjun Zhang
- State Key Laboratory of Oncology in South China, 510060, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China.,Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Kui Wu
- BGI-Shenzhen, 518083, Shenzhen, China. .,China National GeneBank-Shenzhen, BGI-Shenzhen, 518120, Shenzhen, China.
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China. .,State Key Laboratory of Oncology in South China, 510060, Guangzhou, China. .,Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China.
| |
Collapse
|
379
|
Abstract
Large-scale sequencing of human tumours has uncovered a vast array of genomic alterations. Genetically engineered mouse models recapitulate many features of human cancer and have been instrumental in assigning biological meaning to specific cancer-associated alterations. However, their time, cost and labour-intensive nature limits their broad utility; thus, the functional importance of the majority of genomic aberrations in cancer remains unknown. Recent advances have accelerated the functional interrogation of cancer-associated alterations within in vivo models. Specifically, the past few years have seen the emergence of CRISPR-Cas9-based strategies to rapidly generate increasingly complex somatic alterations and the development of multiplexed and quantitative approaches to ascertain gene function in vivo.
Collapse
|
380
|
Fujino T, Kobayashi Y, Suda K, Koga T, Nishino M, Ohara S, Chiba M, Shimoji M, Tomizawa K, Takemoto T, Mitsudomi T. Sensitivity and Resistance of MET Exon 14 Mutations in Lung Cancer to Eight MET Tyrosine Kinase Inhibitors In Vitro. J Thorac Oncol 2019; 14:1753-1765. [PMID: 31279006 DOI: 10.1016/j.jtho.2019.06.023] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/26/2019] [Accepted: 06/26/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND MNNG HOS transforming gene (MET) exon 14 mutations in lung cancer, including exon 14 skipping and point mutations, have been attracting the attention of thoracic oncologists as new therapeutic targets. Tumors with these mutations almost always acquire resistance, which also occurs in other oncogene-addicted lung cancers. However, the resistance mechanisms and treatment strategies are not fully understood. METHODS We generated Ba/F3 cells expressing MET exon 14 mutations by retroviral gene transfer. The sensitivities of these cells to eight MET-tyrosine kinase inhibitors (TKIs) were determined using a colorimetric assay. In addition, using N-ethyl-N-nitrosourea mutagenesis, we generated resistant clones, searched for secondary MET mutations, and then examined the sensitivities of these resistant cells to different TKIs. RESULTS Ba/F3 cells transfected with MET mutations grew in the absence of interleukin-3, indicating their oncogenic activity. These cells were sensitive to all MET-TKIs except tivantinib. We identified a variety of secondary mutations. D1228 and Y1230 were common sites for resistance mutations for type I TKIs, which bind the active form of MET, whereas L1195 and F1200 were common sites for type II TKIs, which bind the inactive form. In general, resistance mutations against type I were sensitive to type II, and vice versa. CONCLUSIONS MET-TKIs inhibited the growth of cells with MET exon 14 mutations. We also identified mutation sites specific for TKI types as resistance mechanisms and complementary activities between type I and type II inhibitors against those mutations. These finding should provide relevant clinical implication for treating patients with lung cancer harboring MET exon 14 mutations.
Collapse
Affiliation(s)
- Toshio Fujino
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yoshihisa Kobayashi
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenichi Suda
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takamasa Koga
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masaya Nishino
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Shuta Ohara
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masato Chiba
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masaki Shimoji
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenji Tomizawa
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Toshiki Takemoto
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tetsuya Mitsudomi
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan.
| |
Collapse
|
381
|
Saigi M, Alburquerque-Bejar JJ, Sanchez-Cespedes M. Determinants of immunological evasion and immunocheckpoint inhibition response in non-small cell lung cancer: the genetic front. Oncogene 2019; 38:5921-5932. [PMID: 31253869 DOI: 10.1038/s41388-019-0855-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
The incorporation into clinical practice of immune-checkpoint inhibitors (ICIs), such as those targeting the cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death 1 (PD-1) and its ligand (PD-L1), has represented a major breakthrough in non-small cell lung cancer (NSCLC) treatment, especially in cases where the cancer has no druggable genetic alterations. Despite becoming the standard of care in certain clinical settings, either alone or in combination with chemotherapy, a proportion of patients do not respond while others actually progress during treatment. Therefore, there is a clinical need to identify accurate predictive biomarkers and to develop novel therapeutic strategies based on ICIs. Although they have limitations, the current markers evaluated to select which patients will undergo ICI treatment are the levels of PD-L1 and the tumor mutational burden. In this paper we describe what is currently known about the dynamic interaction between the cancer cell and the immune system during carcinogenesis, with a particular focus on the description of the functions and gene alterations that preclude the host immunoresponse in NSCLC. We emphasize the deleterious gene alterations in components of the major histocompatibility complex (HLA-I or B2M) and of the response to IFNγ (such as JAK2) which are mutually exclusive and can affect up to one fifth of the NSCLCs. The participation of other gene alterations, such as those of common oncogenes and tumor suppressors, and of the epigenetic alterations will also be discussed, in detail. Finally, we discuss the potential use of the tumor's genetic profile to predict sensitivity to ICIs.
Collapse
Affiliation(s)
- Maria Saigi
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Department of Medical Oncology, Catalan Institute of Oncology (ICO), Avda Gran via, 199-203. L'Hospitalet, 08908, Barcelona, Spain
| | - Juan J Alburquerque-Bejar
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Montse Sanchez-Cespedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain. .,Cancer Genomics Group, Josep Carreras Leukemia Research Institute, IJC Building, Campus ICO-Germans Trias i Pujol Ctra de Can Ruti, Camí de les Escoles s/n, Badalona, 08916, Barcelona, Spain.
| |
Collapse
|
382
|
Wang F, Lu JB, Wu XY, Feng YF, Shao Q, An X, Wang HY. Clinical genetic features and related survival implications in patients with surgically resected large-cell lung cancer. Cancer Manag Res 2019; 11:5489-5499. [PMID: 31354355 PMCID: PMC6585161 DOI: 10.2147/cmar.s200263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/10/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Large-cell lung carcinomas (LCLCs) were reclassified by the World Health Organization 2015 criteria. and remain fairly unknown at the molecular level and targeted-therapeutic options. Methods: Data of 184 lung cancer patients were retrieved from clinical records, of which 54 were found to be pathologically diagnosed as LCLC. The genetic alterations EGFR/KRAS/BRAF mutations, MET copy number, and exon 14 mutation, ALK and ROS1 rearrangements, and PDL1 expression were investigated using clinical technologies. The relationship between clinicopathologic and genetic features was analyzed, and the Kaplan–Meier method with log-rank test was used for analyzing patient survival. Results: Major events, including EGFR, KRAS, and BRAF mutations and MET copy-number gain, were found in 5.6%, 16.7%, 1.9%, and 18.5% in LCLC, respectively. No ALK or ROS1 translocation was detected. PDL1 expression in tumor cells and in tumor-infiltrating lymphocytes was observed in 24 (44.4%) and 16 (29.6%) patients. Kaplan–Meier analysis showed that patients with a KRAS mutation had ower 5-year overall survival than those with wild-type KRAS (25.4% vs 47.8%, P=0.028) and that patients with negative PDL1 stained in tumor cells but positive for tumor-infiltrating lymphocytes had significantly favorable overall survival compared to those with solitary and positive PDL1 stained in tumor cells (62.5% vs 20.6%, P=0.044). Conclusion:KRAS mutations and PDL1 expression can predict patient survival and be potential target options in LCLC.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jia-Bin Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Xiao-Yan Wu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Yan-Fen Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qiong Shao
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Xin An
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Hai-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| |
Collapse
|
383
|
Haynes BC, Blidner RA, Cardwell RD, Zeigler R, Gokul S, Thibert JR, Chen L, Fujimoto J, Papadimitrakopoulou VA, Wistuba II, Latham GJ. An Integrated Next-Generation Sequencing System for Analyzing DNA Mutations, Gene Fusions, and RNA Expression in Lung Cancer. Transl Oncol 2019; 12:836-845. [PMID: 30981944 PMCID: PMC6463765 DOI: 10.1016/j.tranon.2019.02.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/21/2019] [Indexed: 12/25/2022] Open
Abstract
We developed and characterized a next-generation sequencing (NGS) technology for streamlined analysis of DNA and RNA using low-input, low-quality cancer specimens. A single-workflow, targeted NGS panel for non-small cell lung cancer (NSCLC) was designed covering 135 RNA and 55 DNA disease-relevant targets. This multiomic panel was used to assess 219 formalin-fixed paraffin-embedded NSCLC surgical resections and core needle biopsies. Mutations and expression phenotypes were identified consistent with previous large-scale genomic studies, including mutually exclusive DNA and RNA oncogenic driver events. Evaluation of a second cohort of low cell count fine-needle aspirate smears from the BATTLE-2 trial yielded 97% agreement with an independent, validated NGS panel that was used with matched surgical specimens. Collectively, our data indicate that broad, clinically actionable insights that previously required independent assays, workflows, and analyses to assess both DNA and RNA can be conjoined in a first-tier, highly multiplexed NGS test, thereby providing faster, simpler, and more economical results.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vassiliki A Papadimitrakopoulou
- Department of Thoracic/Head and Neck Medical Oncology, Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
384
|
Coomer AO, Black F, Greystoke A, Munkley J, Elliott DJ. Alternative splicing in lung cancer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:194388. [PMID: 31152916 DOI: 10.1016/j.bbagrm.2019.05.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/20/2019] [Indexed: 12/21/2022]
Abstract
Lung cancer has the highest mortality rate of all cancers worldwide. Lung cancer is a very heterogeneous disease that is often diagnosed at later stages which have a poor prognosis. Aberrant alternative splicing patterns found in lung cancer contribute to important cell functions. These include changes in splicing for the BCL2L1, MDM2, MDM4, NUMB and MET genes during lung tumourigenesis, to affect pathways involved in apoptosis, cell proliferation and cellular cohesion. Global analyses of RNASeq datasets suggest there may be many more potentially influential aberrant splicing events that need to be investigated in lung cancer. Changes in expression of the splicing factors that regulate alternative splicing events have also been identified in lung cancer. Of these, changes in expression of QKI, RBM4, RBM5, RBM6, RBM10 and SRSF1 proteins regulate many of the most frequently referenced aberrant splicing events in lung cancer. The expanding list of genes known to be aberrantly spliced in lung cancer along with the altered expression of splicing factors that regulate them are providing new clues as to how lung cancer develops, and how these events can be exploited for better treatment. This article is part of a Special Issue entitled: RNA structure and splicing regulation edited by Francisco Baralle, Ravindra Singh and Stefan Stamm.
Collapse
Affiliation(s)
- Alice O Coomer
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, United Kingdom of Great Britain and Northern Ireland.
| | - Fiona Black
- Cellular Pathology Department, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom of Great Britain and Northern Ireland
| | - Alastair Greystoke
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom of Great Britain and Northern Ireland
| | - Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, United Kingdom of Great Britain and Northern Ireland
| | - David J Elliott
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
385
|
Parizadeh SM, Jafarzadeh-Esfehani R, Fazilat-Panah D, Hassanian SM, Shahidsales S, Khazaei M, Parizadeh SMR, Ghayour-Mobarhan M, Ferns GA, Avan A. The potential therapeutic and prognostic impacts of the c-MET/HGF signaling pathway in colorectal cancer. IUBMB Life 2019; 71:802-811. [PMID: 31116909 DOI: 10.1002/iub.2063] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/10/2019] [Indexed: 12/22/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer and a common cause of cancer-related mortality globally. In spite of the improvements in the early diagnosis of CRC, approximately one-third of patients develop metastasis and then have a very poor survival rate. The mesenchymal-epithelial transition factor (c-MET) is a tyrosine kinase cell surface receptor activated by hepatocyte growth factor (HGF). Activation of c-MET/HGF signaling pathway regulates a variety of biological processes including cell motility, cell proliferation, angiogenesis, the epithelial-to-mesenchymal transition, and the development and progression of cancer cells. Recent studies have suggested that the c-MET/HGF signaling pathway is involved in the carcinogenesis of CRC. In this review, we summarize the main findings of recent studies investigating the role of c-MET/HGF signaling pathway in CRC and the potential of the c-MET/HGF signaling pathways in the diagnosis and treatment of CRC. © 2019 IUBMB Life, 2019.
Collapse
Affiliation(s)
| | - Reza Jafarzadeh-Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Mahdi Hassanian
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Majid Khazaei
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Majid Ghayour-Mobarhan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
386
|
Awad MM, Leonardi GC, Kravets S, Dahlberg SE, Drilon A, Noonan SA, Camidge DR, Ou SHI, Costa DB, Gadgeel SM, Steuer CE, Forde PM, Zhu VW, Fukuda Y, Clark JW, Jänne PA, Mok T, Sholl LM, Heist RS. Impact of MET inhibitors on survival among patients with non-small cell lung cancer harboring MET exon 14 mutations: a retrospective analysis. Lung Cancer 2019; 133:96-102. [PMID: 31200835 DOI: 10.1016/j.lungcan.2019.05.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Although dramatic responses to MET inhibitors have been reported in patients with MET exon 14 (METex14) mutant non-small cell lung cancer (NSCLC), the impact of these treatments on overall survival in this population is unknown. METHODS We conducted a multicenter retrospective analysis of patients with METex14 NSCLC to determine if treatment with MET inhibitors impacts median overall survival (mOS). Event-time distributions were estimated using the Kaplan-Meier method and compared with the log-rank test. Multivariable Cox models were fitted to estimate hazard ratios. RESULTS We identified 148 patients with METex14 NSCLC; the median age was 72; 57% were women and 39% were never smokers. Of the 34 metastatic patients who never received a MET inhibitor, the mOS was 8.1 months; those in this group with concurrent MET amplification had a trend toward worse survival compared to cancers without MET amplification (5.2 months vs 10.5 months, P = 0.06). Of the 27 metastatic patients who received at least one MET inhibitor the mOS was 24.6 months. A model adjusting for receipt of a MET inhibitor as first- or second-line therapy as a time-dependent covariate demonstrated that treatment with a MET inhibitor was associated with a significant prolongation in survival (HR 0.11, 95% CI 0.01-0.92, P = 0.04) compared to patients who did not receive any MET inhibitor. Among 22 patients treated with crizotinib, the median progression-free survival was 7.4 months. DISCUSSION For patients with METex14 NSCLC, treatment with a MET inhibitor is associated with an improvement in overall survival.
Collapse
Affiliation(s)
- Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, USA.
| | - Giulia C Leonardi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Sasha Kravets
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Suzanne E Dahlberg
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, USA
| | | | | | | | - Sai-Hong I Ou
- University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange, USA
| | - Daniel B Costa
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | | | | | - Patrick M Forde
- Johns Hopkins Kimmel Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, USA
| | - Viola W Zhu
- University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange, USA; University of California San Francisco, Fresno, USA
| | | | | | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Tony Mok
- Chinese University of Hong Kong, Hong Kong, China
| | | | | |
Collapse
|
387
|
de Fraipont F, Gazzeri S, Cho WC, Eymin B. Circular RNAs and RNA Splice Variants as Biomarkers for Prognosis and Therapeutic Response in the Liquid Biopsies of Lung Cancer Patients. Front Genet 2019; 10:390. [PMID: 31134126 PMCID: PMC6514155 DOI: 10.3389/fgene.2019.00390] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/10/2019] [Indexed: 01/08/2023] Open
Abstract
Lung cancer, including non-small cell lung carcinoma (NSCLC), is the most frequently diagnosed cancer. It is also the leading cause of cancer-related mortality worldwide because of its late diagnosis and its resistance to therapies. Therefore, the identification of biomarkers for early diagnosis, prognosis, and monitoring of therapeutic response is urgently needed. Liquid biopsies, especially blood, are considered as promising tools to detect and quantify circulating cancer biomarkers. Cell-free circulating tumor DNA has been extensively studied. Recently, the possibility to detect and quantify RNAs in tumor biopsies, notably circulating cell-free RNAs, has gained great attention. RNA alternative splicing contributes to the proteome diversity through the biogenesis of several mRNA splice variants from the same pre-mRNA. Circular RNA (circRNA) is a new class of RNAs resulting from pre-mRNA back splicing. Owing to the development of high-throughput transcriptomic analyses, numerous RNA splice variants and, more recently, circRNAs have been identified and found to be differentially expressed in tumor patients compared to healthy controls. The contribution of some of these RNA splice variants and circRNAs to tumor progression, dissemination, or drug response has been clearly demonstrated in preclinical models. In this review, we discuss the potential of circRNAs and mRNA splice variants as candidate biomarkers for the prognosis and the therapeutic response of NSCLC in liquid biopsies.
Collapse
Affiliation(s)
- Florence de Fraipont
- INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
- Grenoble Hospital, La Tronche, France
| | - Sylvie Gazzeri
- INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Beatrice Eymin
- INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| |
Collapse
|
388
|
Castiglione R, Alidousty C, Holz B, Wagener S, Baar T, Heydt C, Binot E, Zupp S, Kron A, Wolf J, Merkelbach-Bruse S, Reinhardt HC, Buettner R, Schultheis AM. Comparison of the genomic background of MET-altered carcinomas of the lung: biological differences and analogies. Mod Pathol 2019; 32:627-638. [PMID: 30459450 PMCID: PMC6760650 DOI: 10.1038/s41379-018-0182-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 12/22/2022]
Abstract
Although non-small-cell lung cancer is a leading cause of cancer-related deaths, the molecular characterization and classification of its genetic alterations has drastically changed treatment options and overall survival within the last few decades. In particular, tyrosine kinase inhibitors targeting specific molecular alterations, among other MET, have greatly improved the prognosis of non-small-cell lung cancer patients. Here, we compare the genomic background of a subset of non-small-cell lung cancer cases harboring either a MET high-level amplification (n = 24) or a MET exon 14 skipping mutation (n = 26), using next-generatison sequencing, fluorescence in situ hybridization, immunohistochemistry, and Nanostring nCounter® technology. We demonstrate that the MET-amplified cohort shows a higher genetic instability, compared with the mutant cohort (p < 0.001). Furthermore, MET mutations occur at high allele frequency and in the presence of co-occurring TP53 mutations (n = 7), as well as MDM2 (n = 7), CDK4 (n = 6), and HMGA2 (n = 5) co-amplifications. No other potential driver mutation has been detected. Conversely, in the MET-amplified group, we identify co-occurring pathogenic NRAS and KRAS mutations (n = 5) and a significantly higher number of TP53 mutations, compared with the MET-mutant cohort (p = 0.048). Of note, MET amplifications occur more frequently as subclonal events. Interestingly, despite the significantly (p = 0.00103) older age at diagnosis of stage IIIb/IV of MET-mutant patients (median 77 years), compared with MET high-level amplified patients (median 69 years), MET-mutant patients with advanced-stage tumors showed a significantly better prognosis at 12 months (p = 0.04). In conclusion, the two groups of MET genetic alterations differ, both clinically and genetically: our data strongly suggest that MET exon 14 skipping mutations represent an early driver mutation. In opposition, MET amplifications occur usually in the background of other strong genetic events and therefore MET amplifications should be interpreted in the context of each tumor's genetic background, rather than as an isolated driver event, especially when considering MET-specific treatment options.
Collapse
Affiliation(s)
- Roberta Castiglione
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
- Else Kröner Forschungskolleg Clonal Evolution in Cancer, University Hospital Cologne, Cologne, Germany
| | | | - Barbara Holz
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Svenja Wagener
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Till Baar
- Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| | - Carina Heydt
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Elke Binot
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Susann Zupp
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Anna Kron
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Jürgen Wolf
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | | | - Hans Christian Reinhardt
- Else Kröner Forschungskolleg Clonal Evolution in Cancer, University Hospital Cologne, Cologne, Germany
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine, University Hospital of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine, University Hospital of Cologne, Cologne, Germany
| | | |
Collapse
|
389
|
Zhang C, Leighl NB, Wu YL, Zhong WZ. Emerging therapies for non-small cell lung cancer. J Hematol Oncol 2019; 12:45. [PMID: 31023335 PMCID: PMC6482588 DOI: 10.1186/s13045-019-0731-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/05/2019] [Indexed: 02/08/2023] Open
Abstract
Recent advances in the field of novel anticancer agents prolong patients' survival and show a promising future. Tyrosine kinase inhibitors and immunotherapy for lung cancer are the two major areas undergoing rapid development. Although increasing novel anticancer agents were innovated, how to translate and optimize these novel agents into clinical practice remains to be explored. Besides, toxicities and availability of these drugs in specific regions should also be considered during clinical determination. Herein, we summarize emerging agents including tyrosine kinase inhibitors, checkpoint inhibitors, and other potential immunotherapy such as chimeric antigen receptor T cell for non-small cell lung cancer attempting to provide insights and perspectives of the future in anticancer treatment.
Collapse
Affiliation(s)
- Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | | | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
390
|
Qiu Y, Jiang H, Ching WK, Ng MK. On predicting epithelial mesenchymal transition by integrating RNA-binding proteins and correlation data via L1/2-regularization method. Artif Intell Med 2019; 95:96-103. [DOI: 10.1016/j.artmed.2018.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 01/06/2023]
|
391
|
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
|
392
|
Mutation patterns in a population-based non-small cell lung cancer cohort and prognostic impact of concomitant mutations in KRAS and TP53 or STK11. Lung Cancer 2019; 130:50-58. [DOI: 10.1016/j.lungcan.2019.01.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 11/22/2022]
|
393
|
Kohsaka S, Tatsuno K, Ueno T, Nagano M, Shinozaki-Ushiku A, Ushiku T, Takai D, Ikegami M, Kobayashi H, Kage H, Ando M, Hata K, Ueda H, Yamamoto S, Kojima S, Oseto K, Akaike K, Suehara Y, Hayashi T, Saito T, Takahashi F, Takahashi K, Takamochi K, Suzuki K, Nagayama S, Oda Y, Mimori K, Ishihara S, Yatomi Y, Nagase T, Nakajima J, Tanaka S, Fukayama M, Oda K, Nangaku M, Miyazono K, Miyagawa K, Aburatani H, Mano H. Comprehensive assay for the molecular profiling of cancer by target enrichment from formalin-fixed paraffin-embedded specimens. Cancer Sci 2019; 110:1464-1479. [PMID: 30737998 PMCID: PMC6447855 DOI: 10.1111/cas.13968] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/17/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
Tumor molecular profiling is becoming a standard of care for patients with cancer, but the optimal platform for cancer sequencing remains undetermined. We established a comprehensive assay, the Todai OncoPanel (TOP), which consists of DNA and RNA hybridization capture‐based next‐generation sequencing panels. A novel method for target enrichment, named the junction capture method, was developed for the RNA panel to accurately and cost‐effectively detect 365 fusion genes as well as aberrantly spliced transcripts. The TOP RNA panel can also measure the expression profiles of an additional 109 genes. The TOP DNA panel was developed to detect single nucleotide variants and insertions/deletions for 464 genes, to calculate tumor mutation burden and microsatellite instability status, and to infer chromosomal copy number. Clinically relevant somatic mutations were identified in 32.2% (59/183) of patients by prospective TOP testing, signifying the clinical utility of TOP for providing personalized medicine to cancer patients.
Collapse
Affiliation(s)
- Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenji Tatsuno
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Masaaki Nagano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Shinozaki-Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daiya Takai
- Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Masachika Ikegami
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Orthopedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Kobayashi
- Department of Orthopedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mizuo Ando
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keisuke Hata
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki Ueda
- Biological Data Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shogo Yamamoto
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shinya Kojima
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kumiko Oseto
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Keisuke Akaike
- Department of Orthopedic Surgery, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Orthopedic Surgery, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University, Graduate School of Medicine, Tokyo, Japan.,Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Nagayama
- Gastroenterological Center, Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Soichiro Ishihara
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaomi Nangaku
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
394
|
Blidner RA, Haynes BC, Hyter S, Schmitt S, Pessetto ZY, Godwin AK, Su D, Hurban P, van Kempen LC, Aguirre ML, Gokul S, Cardwell RD, Latham GJ. Design, Optimization, and Multisite Evaluation of a Targeted Next-Generation Sequencing Assay System for Chimeric RNAs from Gene Fusions and Exon-Skipping Events in Non-Small Cell Lung Cancer. J Mol Diagn 2019; 21:352-365. [PMID: 30529127 PMCID: PMC7057224 DOI: 10.1016/j.jmoldx.2018.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/11/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Lung cancer accounts for approximately 14% of all newly diagnosed cancers and is the leading cause of cancer-related deaths. Chimeric RNA resulting from gene fusions (RNA fusions) and other RNA splicing errors are driver events and clinically addressable targets for non-small cell lung cancer (NSCLC). The reliable assessment of these RNA markers by next-generation sequencing requires integrated reagents, protocols, and interpretive software that can harmonize procedures and ensure consistent results across laboratories. We describe the development and verification of a system for targeted RNA sequencing for the analysis of challenging, low-input solid tumor biopsies that includes reagents for nucleic acid quantification and library preparation, run controls, and companion bioinformatics software. Assay development reconciled sequence discrepancies in public databases, created predictive formalin-fixed, paraffin-embedded RNA qualification metrics, and eliminated read misidentification attributable to index hopping events on the next-generation sequencing flow cell. The optimized and standardized system was analytically verified internally and in a multiphase study conducted at five independent laboratories. The results show accurate, reproducible, and sensitive detection of RNA fusions, alternative splicing events, and other expression markers of NSCLC. This comprehensive approach, combining sample quantification, quality control, library preparation, and interpretive bioinformatics software, may accelerate the routine implementation of targeted RNA sequencing of formalin-fixed, paraffin-embedded samples relevant to NSCLC.
Collapse
Affiliation(s)
| | | | - Stephen Hyter
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sarah Schmitt
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Ziyan Y Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas; University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas
| | - Dan Su
- Q Squared Solutions Expression Analysis LLC, Morrisville, North Carolina
| | - Patrick Hurban
- Q Squared Solutions Expression Analysis LLC, Morrisville, North Carolina
| | - Léon C van Kempen
- The Molecular Pathology Centre, Jewish General Hospital, Montreal, Quebec, Canada
| | - Maria L Aguirre
- The Molecular Pathology Centre, Jewish General Hospital, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
395
|
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
|
396
|
Robinson TJ, Freedman JA, Al Abo M, Deveaux AE, LaCroix B, Patierno BM, George DJ, Patierno SR. Alternative RNA Splicing as a Potential Major Source of Untapped Molecular Targets in Precision Oncology and Cancer Disparities. Clin Cancer Res 2019; 25:2963-2968. [PMID: 30755441 PMCID: PMC6653604 DOI: 10.1158/1078-0432.ccr-18-2445] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/18/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022]
Abstract
Studies of alternative RNA splicing (ARS) have the potential to provide an abundance of novel targets for development of new biomarkers and therapeutics in oncology, which will be necessary to improve outcomes for patients with cancer and mitigate cancer disparities. ARS, a key step in gene expression enabling individual genes to encode multiple proteins, is emerging as a major driver of abnormal phenotypic heterogeneity. Recent studies have begun to identify RNA splicing-related genetic and genomic variation in tumors, oncogenes dysregulated by ARS, RNA splice variants driving race-related cancer aggressiveness and drug response, spliceosome-dependent transformation, and RNA splicing-related immunogenic epitopes in cancer. In addition, recent studies have begun to identify and test, preclinically and clinically, approaches to modulate and exploit ARS for therapeutic application, including splice-switching oligonucleotides, small molecules targeting RNA splicing or RNA splice variants, and combination regimens with immunotherapies. Although ARS data hold such promise for precision oncology, inclusion of studies of ARS in translational and clinical cancer research remains limited. Technologic developments in sequencing and bioinformatics are being routinely incorporated into clinical oncology that permit investigation of clinically relevant ARS events, yet ARS remains largely overlooked either because of a lack of awareness within the clinical oncology community or perceived barriers to the technical complexity of analyzing ARS. This perspective aims to increase such awareness, propose immediate opportunities to improve identification and analysis of ARS, and call for bioinformaticians and cancer researchers to work together to address the urgent need to incorporate ARS into cancer biology and precision oncology.
Collapse
Affiliation(s)
| | - Jennifer A Freedman
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Muthana Al Abo
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - April E Deveaux
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Bonnie LaCroix
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Brendon M Patierno
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Daniel J George
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Steven R Patierno
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina.
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
397
|
Grosse A, Grosse C, Rechsteiner M, Soltermann A. Analysis of the frequency of oncogenic driver mutations and correlation with clinicopathological characteristics in patients with lung adenocarcinoma from Northeastern Switzerland. Diagn Pathol 2019; 14:18. [PMID: 30744664 PMCID: PMC6371584 DOI: 10.1186/s13000-019-0789-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/01/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Molecular testing of lung adenocarcinoma for oncogenic driver mutations has become standard in pathology practice. The aim of the study was to analyze the EGFR, KRAS, ALK, RET, ROS1, BRAF, ERBB2, MET and PIK3CA mutational status in a representative cohort of Swiss patients with lung adenocarcinoma and to correlate the mutational status with clinicopathological patient characteristics. METHODS All patients who underwent molecular testing of newly diagnosed lung adenocarcinoma during a 4-year period (2014-2018) were included. Molecular analyses were performed with Sanger sequencing (n = 158) and next generation sequencing (n = 311). ALK, ROS1 and RET fusion gene analyses were also performed with fluorescence in situ hybridization and immunohistochemistry/immunocytochemistry. Demographic and clinical data were obtained from the medical records. RESULTS Of 469 patients with informative EGFR mutation analyses, 90 (19.2%) had EGFR mutations. KRAS mutations were present in 33.9% of the patients, while 6.0% of patients showed ALK rearrangement. BRAF, ERBB2, MET and PIK3CA mutations and ROS1 and RET rearrangements were found in 2.6%, 1.9%, 1.9%, 1.5%, 1.7% and 0.8% of the patients, respectively. EGFR mutation was significantly associated with female gender and never smoking status. ALK translocations were more frequent in never smokers, while KRAS mutations were more commonly found in ever smokers. The association between KRAS mutational status and female gender was statistically significant only on multivariate analysis after adjusting for smoking. CONCLUSION The EGFR mutation rate in the current study is among the higher previously reported mutation rates, while the frequencies of KRAS, BRAF, ERBB2 and PIK3CA mutations and ALK, ROS1 and RET rearrangements are similar to the results of previous reports. EGFR and KRAS mutations were significantly associated with gender and smoking. ALK rearrangements showed a significant association with smoking status alone.
Collapse
Affiliation(s)
- Alexandra Grosse
- Institute of Pathology and Molecular Pathology, Clinical Pathology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Claudia Grosse
- Institute of Pathology, Kepler University Hospital, Krankenhausstraße 9, 4021 Linz, Austria
| | - Markus Rechsteiner
- Institute of Pathology and Molecular Pathology, Diagnostic Molecular Pathology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Alex Soltermann
- Institute of Pathology and Molecular Pathology, Clinical Pathology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
398
|
Teixidó C, Giménez-Capitán A, Molina-Vila MÁ, Peg V, Karachaliou N, Rodríguez-Capote A, Castellví J, Rosell R. RNA Analysis as a Tool to Determine Clinically Relevant Gene Fusions and Splice Variants. Arch Pathol Lab Med 2019; 142:474-479. [PMID: 29565207 DOI: 10.5858/arpa.2017-0134-ra] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - Technologic advances have contributed to the increasing relevance of RNA analysis in clinical oncology practice. The different genetic aberrations that can be screened with RNA include gene fusions and splice variants. Validated methods of identifying these alterations include fluorescence in situ hybridization, immunohistochemistry, reverse transcription-polymerase chain reaction, and next-generation sequencing, which can provide physicians valuable information on disease and treatment of cancer patients. OBJECTIVE - To discuss the standard techniques available and new approaches for the identification of gene fusions and splice variants in cancer, focusing on RNA analysis and how analytic methods have evolved in both tissue and liquid biopsies. DATA SOURCES - This is a narrative review based on PubMed searches and the authors' own experiences. CONCLUSIONS - Reliable RNA-based testing in tissue and liquid biopsies can inform the diagnostic process and guide physicians toward the best treatment options. Next-generation sequencing methodologies permit simultaneous assessment of molecular alterations and increase the number of treatment options available for cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rafael Rosell
- From the Department of Pathology, Hospital Clínic, Barcelona, Spain (Dr Teixidó); Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (Dr Teixidó); Pangaea Oncology, Oncology Laboratory, Dexeus University Hospital - Quirónsalud Group, Barcelona, Spain (Ms Giménez-Capitán and Drs Molina-Vila, Peg, Karachaliou, Castellví, and Rosell); the Department of Pathology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (Drs Peg and Castellví); Morphological Sciences Department, Universitat Autònoma de Barcelona, Barcelona, Spain (Drs Peg and Castellví); Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor and Quirónsalud Group, Barcelona, Spain (Drs Karachaliou and Rosell); the Department of Medical Oncology, Canarias University Hospital, San Cristóbal de La Laguna, Tenerife, Spain (Dr Rodríguez-Capote); and Cancer Biology & Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Badalona, Spain (Dr Rosell)
| |
Collapse
|
399
|
Baltschukat S, Engstler BS, Huang A, Hao HX, Tam A, Wang HQ, Liang J, DiMare MT, Bhang HEC, Wang Y, Furet P, Sellers WR, Hofmann F, Schoepfer J, Tiedt R. Capmatinib (INC280) Is Active Against Models of Non–Small Cell Lung Cancer and Other Cancer Types with Defined Mechanisms of MET Activation. Clin Cancer Res 2019; 25:3164-3175. [DOI: 10.1158/1078-0432.ccr-18-2814] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/12/2018] [Accepted: 01/18/2019] [Indexed: 11/16/2022]
|
400
|
O'Brien O, Wright MC, O'Brien C, Geoghegan O, Leonard N, Nicholson S, Cuffe S, Fabre A, Jochum W, Joerger M, Gray SG, Finn SP. Cost-Efficient and Easy to Perform PCR-Based Assay to Identify Met Exon 14 Skipping in Formalin-Fixed Paraffin-Embedded (FFPE) Non-Small Cell Lung Cancer (NSCLC) Samples. Diagnostics (Basel) 2019; 9:diagnostics9010013. [PMID: 30669306 PMCID: PMC6468531 DOI: 10.3390/diagnostics9010013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/10/2019] [Accepted: 01/14/2019] [Indexed: 01/03/2023] Open
Abstract
MET is a receptor tyrosine kinase (RTK) that plays important roles in carcinogenesis. Despite being frequently overexpressed in cancer, clinical responses to targeting this receptor have been limited. Recently novel splicing mutations involving the loss of exon 14 (called METex14 skipping) have emerged as potential biomarkers to predict for responsiveness to targeted therapies with Met inhibitors in non-small cell lung cancer (NSCLC). Currently, the diverse genomic alterations responsible for METex14 skipping pose a challenge for routine clinical diagnostic testing. In this report, we examine three different methodologies to detect METex14 and assess their potential utility for use as a diagnostic assay for both the identification of METex14 and intra-tumoural distribution in NSCLC.
Collapse
Affiliation(s)
- Odharnaith O'Brien
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Mark C Wright
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Cathal O'Brien
- Cancer Molecular Diagnostics, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Orla Geoghegan
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
| | - Niamh Leonard
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Siobhan Nicholson
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
| | - Sinéad Cuffe
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- HOPE Directorate, St. James's Hospital, D08 RT2X Dublin, Ireland.
| | - Aurelie Fabre
- Department of Pathology, St. Vincent's University Hospital, University College Dublin School of Medicine, D04 T6F4 Dublin, Ireland.
| | - Wolfram Jochum
- Department of Pathology, Cantonal Hospital, 9007 St. Gallen, Switzerland.
| | - Markus Joerger
- Department of Medical Oncology & Hematology, Cantonal Hospital, 9007 St. Gallen, Switzerland.
| | - Steven G Gray
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- Department of Clinical Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland.
- School of Biological Sciences, Dublin Institute of Technology, D08 NF82 Dublin, Ireland.
| | - Stephen P Finn
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, D08 W9RT Dublin, Ireland.
- Department of Histopathology, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
- Cancer Molecular Diagnostics, Labmed Directorate, St. James's Hospital, D08 RX0X Dublin, Ireland.
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, D08 X4RX Dublin, Ireland.
| |
Collapse
|