251
|
Dagogo-Jack I, Moonsamy P, Gainor JF, Lennerz JK, Piotrowska Z, Lin JJ, Lennes IT, Sequist LV, Shaw AT, Goodwin K, Stevens SE, Do A, Digumarthy SR, Price K, Muzikansky A, Hata AN, Heist RS. A Phase 2 Study of Capmatinib in Patients With MET-Altered Lung Cancer Previously Treated With a MET Inhibitor. J Thorac Oncol 2021; 16:850-859. [PMID: 33545388 DOI: 10.1016/j.jtho.2021.01.1605] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/02/2021] [Accepted: 01/02/2021] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Capmatinib is approved for MET exon 14-altered NSCLC on the basis of activity in targeted therapy-naive patients. We conducted a phase 2 study to assess the efficacy of capmatinib in patients previously treated with a MET inhibitor. METHODS Patients with advanced NSCLC harboring MET amplification or MET exon 14 skipping alterations received capmatinib 400 mg twice daily. The primary end point was the objective response rate. Secondary end points included progression-free survival, disease control rate (DCR), intracranial response rate, and overall survival. Circulating tumor DNA was analyzed to identify capmatinib resistance mechanisms. RESULTS A total of 20 patients were enrolled between May 2016 and November 2019, including 15 patients with MET skipping alterations and five patients with MET amplification. All patients had received crizotinib; three had also received other MET-directed therapies. The median interval between crizotinib and capmatinib was 22 days (range: 4-374). Two patients (10%) achieved an objective response to capmatinib and 14 had stable disease, yielding a DCR of 80%. Among five patients who discontinued crizotinib for intolerance, the DCR was 83%, including two patients with the best tumor shrinkage of -25% and -28%. Intracranial DCR among four patients with measurable brain metastases was 100%, with no observed intracranial objective responses. Overall, the median progression-free survival and overall survival were 5.5 (95% confidence interval: 1.3-11.0) and 11.3 (95% confidence interval: 5.5-not reached) months, respectively. MET D1228 and Y1230 mutations and MAPK alterations were recurrently detected in postcrizotinib, precapmatinib plasma. New and persistent MET mutations and MAPK pathway alterations were detected in plasma at progression on capmatinib. CONCLUSIONS Capmatinib has modest activity in crizotinib-pretreated MET-altered NSCLC, potentially owing to overlapping resistance mechanisms.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Philicia Moonsamy
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Justin F Gainor
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Massachusetts General Hospital, Boston, Massachusetts
| | - Zofia Piotrowska
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Inga T Lennes
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Novartis Institutes of Biomedical Research, Cambridge, Massachusetts
| | - Kelly Goodwin
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Sara E Stevens
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Andrew Do
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Subba R Digumarthy
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Alona Muzikansky
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Rebecca S Heist
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
252
|
He Y, Song L, Wang H, Chen P, Liu Y, Sun H, Li X, Dang S, Liu G, Liu X, Chen S, Zhang X, Hofman P, Uchino J, Park HS, Pacheco JM, Tabbò F, Xu M, Dai J, He K, Yang Y, Zhou C. Mutational Profile Evaluates Response and Survival to First-Line Chemotherapy in Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003263. [PMID: 33643802 PMCID: PMC7887584 DOI: 10.1002/advs.202003263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/04/2020] [Indexed: 05/06/2023]
Abstract
Evaluating the therapeutic response and survival of lung cancer patients receiving first-line chemotherapy has always been difficult. Limited biomarkers for evaluation exist and as a result histology represents an empiric tool to guide therapeutic decision making. In this study, molecular signatures associated with response and long-term survival of lung cancer patients receiving first-line chemotherapy are discovered. Whole-exome sequencing is performed on pretherapeutic tissue samples of 186 patients [145 non-small cell lung cancer (NSCLC) and 41 small cell lung cancer (SCLC)]. On the basis of genomic alteration characteristics, NSCLC patients can be classified into four subtypes (C1-C4). The long-term survival is similar among different subtypes. SCLC patients are also divided into four subtypes and significant difference in their progression free survival is revealed (P < 0.001). NSCLC patients can be divided into three subtypes (S1-S3) based on TMB. A trend of worse survival associated with higher TMB in subtype S3 than in S1+S2 is found. In contrast, no significant correlations between molecular subtype and therapeutic response are observed. In conclusion, this study identifies several molecular signatures associated with response and survival to first-line chemotherapy in lung cancer.
Collapse
Affiliation(s)
- Yayi He
- Department of Medical OncologyShanghai Pulmonary Hospital and Thoracic Cancer InstituteTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | - Lele Song
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
- Department of Radiotherapythe eighth medical center of the Chinese PLA General HospitalNo. 17, Heishanhu Road, Haidian DistrictBeijing100091P. R. China
| | - Hao Wang
- Department of Medical OncologyShanghai Pulmonary Hospital and Thoracic Cancer InstituteTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | - Peixin Chen
- Department of Medical OncologyShanghai Pulmonary Hospital and Thoracic Cancer InstituteTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | - Yu Liu
- Department of Medical OncologyShanghai Pulmonary Hospital and Thoracic Cancer InstituteTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | - Hui Sun
- Department of Medical OncologyShanghai Pulmonary Hospital and Thoracic Cancer InstituteTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | - Xiaobin Li
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Shiying Dang
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Guifeng Liu
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Xinyi Liu
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Shifu Chen
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Xiaoni Zhang
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
- Shenzhen HaploX Medical Laboratory1106 South Block of Yuanxing Science and Technology Building, No. 1 Songpingshan Road, Xili Street, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Paul Hofman
- Laboratory of Clinical and Experimental PathologyFHU OncoAgeBB‐0033‐00025Pasteur HospitalUniversity Côte d'Azur30 avenue de la voie romaine, Nice cedex 01Nice06001France
| | - Junji Uchino
- Department of Pulmonary MedicineKyoto Prefectural University of MedicineKyoto602‐8566Japan
| | - Henry S. Park
- Department of Therapeutic RadiologyYale University School of MedicineNew HavenCT06511USA
| | - Jose M. Pacheco
- Thoracic Oncology ProgramDivision of Medical OncologyDepartment of Internal MedicineUniversity of Colorado Cancer CenterAuroraCO80045USA
| | - Fabrizio Tabbò
- Department of OncologyUniversity of TurinSan Luigi HospitalOrbassanoTurin10043Italy
| | - Mingyan Xu
- HaploX Biotechnology, Co., Ltd.8th floor, Auto Electric Power Building, Songpingshan Road, Nanshan DistrictShenzhenGuangdong518057P. R. China
| | - Jiawei Dai
- SJTU‐Yale Joint Center for Biostatistics and Data ScienceDepartment of Bioinformatics and BiostatisticsSchool of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
| | - Kan He
- SJTU‐Yale Joint Center for Biostatistics and Data ScienceDepartment of Bioinformatics and BiostatisticsSchool of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
| | - Yang Yang
- Department of Surgery, Shanghai Pulmonary HospitalTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | - Caicun Zhou
- Department of Medical OncologyShanghai Pulmonary Hospital and Thoracic Cancer InstituteTongji University School of MedicineNo. 507, Zhengmin Road, Yangpu DistrictShanghai200433P. R. China
| | | |
Collapse
|
253
|
Viñal D, Martínez D, Higuera O, de Castro J. Genomic profiling in non-small-cell lung cancer in young patients. A systematic review. ESMO Open 2021; 6:100045. [PMID: 33516149 PMCID: PMC7844571 DOI: 10.1016/j.esmoop.2020.100045] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer in young patients is an uncommon and understudied entity that harbors distinctive epidemiological, clinic-demographic, and genomic features. We carried out a systematic review of genomic profiling in young patients with lung cancer from 2010 to 2020 in the main electronic databases and selected 23 manuscripts. Lung cancer in young patients occurs more frequently in women with adenocarcinoma histology and at more advanced stages. Some studies report higher oncogenic genomic alteration in this population, with higher anaplastic lymphoma kinase rearrangements, a distinct profile of epidermal growth factor receptor mutations, and other novel genomic alterations. Although still uncommon, the implementation of next-generation sequencing (NGS) has shed some light on germline genomic alterations associated with lung cancer in young patients. Although outcomes when compared with the older population are conflicting, the overall prognosis is still poor in this subset of patients and efforts to find targetable genomic alterations should be made to improve survival.
Collapse
Affiliation(s)
- D Viñal
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain.
| | - D Martínez
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
| | - O Higuera
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
| | - J de Castro
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain; Experimental Therapies and Novel Biomarkers in Cancer IdiPAZ, Madrid, Spain; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC, Madrid, Spain
| |
Collapse
|
254
|
Elsayed M, Christopoulos P. Therapeutic Sequencing in ALK + NSCLC. Pharmaceuticals (Basel) 2021; 14:ph14020080. [PMID: 33494549 PMCID: PMC7912146 DOI: 10.3390/ph14020080] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/17/2022] Open
Abstract
Anaplastic lymphoma kinase-rearranged non-small-cell lung cancer (ALK+ NSCLC) is a model disease for the use of targeted pharmaceuticals in thoracic oncology. Due to higher systemic and intracranial efficacy, the second-generation ALK tyrosine kinase inhibitors (TKI) alectinib and brigatinib have irrevocably displaced crizotinib as standard first-line treatment, based on the results of the ALEX and ALTA-1L trials. Besides, lorlatinib and brigatinib are the preferred second-line therapies for progression under second-generation TKI and crizotinib, respectively, based on the results of several phase II studies. Tissue or liquid rebiopsies at the time of disease progression, even though not mandated by the approval status of any ALK inhibitor, are gaining importance for individualization and optimization of patient management. Of particular interest are cases with off-target resistance, for example MET, HER2 or KRAS alterations, which require special therapeutic maneuvers, e.g., inclusion in early clinical trials or off-label administration of respectively targeted drugs. On the other hand, up to approximately half of the patients failing TKI, develop anatomically restricted progression, which can be initially tackled with local ablative measures without switch of systemic therapy. Among the overall biologically favorable ALK+ tumors, with a mean tumor mutational burden uniquely below 3 mutations per Mb and the longest survival among NSCLC currently, presence of the EML4-ALK fusion variant 3 and/or TP53 mutations identify high-risk cases with earlier treatment failure and a need for more aggressive surveillance and treatment strategies. The potential clinical utility of longitudinal ctDNA assays for earlier detection of disease progression and improved guidance of therapy in these patients is a currently a matter of intense investigation. Major pharmaceutical challenges for the field are the development of more potent, fourth-generation TKI and effective immuno-oncological interventions, especially ALK-directed cell therapies, which will be essential for further improving survival and achieving cure of ALK+ tumors.
Collapse
Affiliation(s)
- Mei Elsayed
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, 69126 Heidelberg, Germany;
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, 69126 Heidelberg, Germany;
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69126 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-396-1371
| |
Collapse
|
255
|
Herbst RS, Aisner DL, Sonett JR, Turk AT, Weintraub JL, Lindeman NI. Practical Considerations Relating to Routine Clinical Biomarker Testing for Non-small Cell Lung Cancer: Focus on Testing for RET Fusions. Front Med (Lausanne) 2021; 7:562480. [PMID: 33553195 PMCID: PMC7859651 DOI: 10.3389/fmed.2020.562480] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/03/2020] [Indexed: 11/15/2022] Open
Abstract
For patients with advanced non–small cell lung cancer, genomic profiling of tumors to identify potentially targetable alterations and thereby inform treatment selection is now part of standard care. While molecular analyses are primarily focused on actionable biomarkers associated with regulatory agency-approved therapies, there are a number of emerging biomarkers linked to investigational agents in advanced stages of clinical development will become approved agents. A particularly timely example is the reported data and US Food and Drug Administration approval of highly specific small molecule inhibitors of the proto-oncogene tyrosine-protein kinase receptor RET indicate that testing for tumor RET gene fusions in patients with NSCLC has become clinically important. As the number of biomarkers to be tested in NSCLC grows, it becomes increasingly important to optimize and prioritize the use of biopsy tissue, in order to both continue to allow accurate histopathological diagnosis and also to support concurrent genomic profiling to identify perhaps relatively uncommon genetic events. In order to provide practical expert consensus guidance to optimize processes facilitating genomic testing in NSCLC and to overcome barriers to access and implementation, a multidisciplinary advisory board was held in New York, on January 30, 2019. The panel comprised physicians involved in sample procurement (interventional radiologists and a thoracic surgeon), surgical pathologists specializing in the lung, molecular pathologists, and thoracic oncologists. Particular consideration was given to the key barriers faced by these experts in establishing institutional genomic screening programs for NSCLC. Potential solutions have been devised in the form of consensus opinions that might be used to help resolve such issues.
Collapse
Affiliation(s)
- Roy S Herbst
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Joshua R Sonett
- Division of Thoracic Surgery, Lung Transplant Program, Columbia University Medical Center, New York, NY, United States
| | - Andrew T Turk
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Joshua L Weintraub
- Division of Interventional Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
256
|
Lu C, Zhou Q. Diagnostics, therapeutics and RET inhibitor resistance for RET fusion-positive non-small cell lung cancers and future perspectives. Cancer Treat Rev 2021; 96:102153. [PMID: 33773204 DOI: 10.1016/j.ctrv.2021.102153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/08/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
Selective RET inhibitors is the current hot topic, making multikinase inhibitors a thing of the past. However, the limitation of various test approaches, coupled with lack of knowledge of acquired resistance mechanisms, and specific patient groups that bear special consideration, remains a challenge. Herein, we outline utility of various diagnostic techniques, provide evidence to guide management of RET-fusion-positive Non-Small Cell Lung Cancer (NSCLC) patients, including specific patient groups, such as EGFR-mutant NSCLC patients who acquired RET fusions after resisting EGFR TKIs, and offer a compendium of mechanisms of acquired resistance to RET targeted therapies. This review further provides a list of ongoing clinical trials and summarizes perspectives to guide future development of drugs and trials for this population.
Collapse
Affiliation(s)
- Chang Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
257
|
Bebb DG, Banerji S, Blais N, Desmeules P, Gill S, Grin A, Feilotter H, Hansen AR, Hyrcza M, Krzyzanowska M, Melosky B, Noujaim J, Purgina B, Ruether D, Simmons CE, Soulieres D, Torlakovic EE, Tsao MS. Canadian Consensus for Biomarker Testing and Treatment of TRK Fusion Cancer in Adults. Curr Oncol 2021; 28:523-548. [PMID: 33467570 PMCID: PMC7903287 DOI: 10.3390/curroncol28010053] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The tyrosine receptor kinase (TRK) inhibitors larotrectinib and entrectinib were recently approved in Canada for the treatment of solid tumours harbouring neurotrophic tyrosine receptor kinase (NTRK) gene fusions. These NTRK gene fusions are oncogenic drivers found in most tumour types at a low frequency (<5%), and at a higher frequency (>80%) in a small number of rare tumours (e.g., secretory carcinoma of the salivary gland and of the breast). They are generally mutually exclusive of other common oncogenic drivers. Larotrectinib and entrectinib have demonstrated impressive overall response rates and tolerability in Phase I/II trials in patients with TRK fusion cancer with no other effective treatment options. Given the low frequency of TRK fusion cancer and the heterogeneous molecular testing landscape in Canada, identifying and optimally managing such patients represents a new challenge. We provide a Canadian consensus on when and how to test for NTRK gene fusions and when to consider treatment with a TRK inhibitor. We focus on five tumour types: thyroid carcinoma, colorectal carcinoma, non-small cell lung carcinoma, soft tissue sarcoma, and salivary gland carcinoma. Based on the probability of the tumour harbouring an NTRK gene fusion, we also suggest a tumour-agnostic consensus for NTRK gene fusion testing and treatment. We recommend considering a TRK inhibitor in all patients with TRK fusion cancer with no other effective treatment options.
Collapse
Affiliation(s)
- D. Gwyn Bebb
- Tom Baker Cancer Centre and University of Calgary, Calgary, AB T2N 4N2, Canada
| | - Shantanu Banerji
- Research Institute in Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Normand Blais
- Centre Hospitalier Universitaire de Montreal, Department of Medicine, University of Montreal, Montreal, QC H2X 3E4, Canada; (N.B.); (D.S.)
| | - Patrice Desmeules
- Service D’Anatomopathologie et de Cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC G1V 0A6, Canada;
| | - Sharlene Gill
- BC Cancer, Vancouver, BC V5Z 4E6, Canada; (S.G.); (B.M.); (C.E.S.)
| | - Andrea Grin
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (H.F.)
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (H.F.)
| | - Aaron R. Hansen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (A.R.H.); (M.K.)
| | - Martin Hyrcza
- Department of Pathology and Laboratory Medicine, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada;
| | - Monika Krzyzanowska
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (A.R.H.); (M.K.)
| | - Barbara Melosky
- BC Cancer, Vancouver, BC V5Z 4E6, Canada; (S.G.); (B.M.); (C.E.S.)
| | | | - Bibiana Purgina
- The Ottawa Hospital, Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Dean Ruether
- Department of Oncology, Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada;
| | | | - Denis Soulieres
- Centre Hospitalier Universitaire de Montreal, Department of Medicine, University of Montreal, Montreal, QC H2X 3E4, Canada; (N.B.); (D.S.)
| | - Emina Emilia Torlakovic
- Department of Pathology and Laboratory Medicine, Saskatchewan Health Authority and University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | - Ming-Sound Tsao
- Department of Pathology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
258
|
Fusco MJ, Saeed-Vafa D, Carballido EM, Boyle TA, Malafa M, Blue KL, Teer JK, Walko CM, McLeod HL, Hicks JK, Extermann M, Fleming JB, Knepper TC, Kim DW. Identification of Targetable Gene Fusions and Structural Rearrangements to Foster Precision Medicine in KRAS Wild-Type Pancreatic Cancer. JCO Precis Oncol 2021; 5:PO.20.00265. [PMID: 34250383 PMCID: PMC8232071 DOI: 10.1200/po.20.00265] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023] Open
Abstract
It has recently been described that alternative oncogenic drivers may be found in KRAS wild-type (KRAS WT) pancreatic cancers. This study aimed to determine the incidence of targetable gene fusions present in KRAS WT pancreatic adenocarcinoma and response to targeted therapy. METHODS One hundred consecutive patients with pancreatic adenocarcinoma who underwent targeted next-generation sequencing using DNA sequencing with RNA sequencing (n = 47) or without RNA sequencing (n = 53) at a single institution were included in the study. The frequency and landscape of targetable fusions in KRAS WT pancreatic adenocarcinoma was characterized and compared with the frequency of fusions in KRAS-mutated (KRAS MUT) pancreatic adenocarcinoma. Results were validated in two independent cohorts using data from AACR GENIE (n = 1,252) and TCGA (n = 150). The clinical history of fusion-positive patients who received targeted treatment is described. RESULTS Pancreatic cancers from 13 of 100 patients (13%) were found to be KRAS WT. Targetable fusions were identified in 4/13 (31%) KRAS WT tumors compared with 0/87 (0%) KRAS MUT pancreatic adenocarcinomas (P = .0002). One patient with a novel MET fusion had a complete response to targeted therapy with crizotinib that is ongoing at 12+ months of treatment. In the validation cohorts, gene fusions were identified in 18/97 (19%) and 2/10 (20%) KRAS WT tumors reported in the AACR GENIE and TCGA cohorts, respectively. CONCLUSION Oncogene fusions are present in KRAS WT pancreatic adenocarcinomas at an increased frequency when compared with KRAS MUT pancreatic adenocarcinomas. As these fusions may be susceptible to targeted therapy, molecular analyses for the detection of fusions in KRAS WT pancreatic adenocarcinomas may warrant increased consideration.
Collapse
Affiliation(s)
- Michael J. Fusco
- Department of Individualized Cancer Management Section of Precision Oncology, Moffitt Cancer Center, Tampa, FL
| | | | | | - Theresa A. Boyle
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL
| | - Kirsten L. Blue
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL
| | - Jamie K. Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL
| | - Christine M. Walko
- Department of Individualized Cancer Management Section of Precision Oncology, Moffitt Cancer Center, Tampa, FL
| | - Howard L. McLeod
- Geriatric Oncology Consortium, Tampa, FL
- USF Taneja College of Pharmacy, Tampa, FL
| | - J. Kevin Hicks
- Department of Individualized Cancer Management Section of Precision Oncology, Moffitt Cancer Center, Tampa, FL
| | - Martine Extermann
- Department of Senior Adult Oncology, Moffitt Cancer Center, Tampa, FL
| | - Jason B. Fleming
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL
| | - Todd C. Knepper
- Department of Individualized Cancer Management Section of Precision Oncology, Moffitt Cancer Center, Tampa, FL
| | - Dae Won Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
259
|
Drilon A, Jenkins C, Iyer S, Schoenfeld A, Keddy C, Davare MA. ROS1-dependent cancers - biology, diagnostics and therapeutics. Nat Rev Clin Oncol 2021; 18:35-55. [PMID: 32760015 PMCID: PMC8830365 DOI: 10.1038/s41571-020-0408-9] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
Abstract
The proto-oncogene ROS1 encodes a receptor tyrosine kinase with an unknown physiological role in humans. Somatic chromosomal fusions involving ROS1 produce chimeric oncoproteins that drive a diverse range of cancers in adult and paediatric patients. ROS1-directed tyrosine kinase inhibitors (TKIs) are therapeutically active against these cancers, although only early-generation multikinase inhibitors have been granted regulatory approval, specifically for the treatment of ROS1 fusion-positive non-small-cell lung cancers; histology-agnostic approvals have yet to be granted. Intrinsic or extrinsic mechanisms of resistance to ROS1 TKIs can emerge in patients. Potential factors that influence resistance acquisition include the subcellular localization of the particular ROS1 oncoprotein and the TKI properties such as the preferential kinase conformation engaged and the spectrum of targets beyond ROS1. Importantly, the polyclonal nature of resistance remains underexplored. Higher-affinity next-generation ROS1 TKIs developed to have improved intracranial activity and to mitigate ROS1-intrinsic resistance mechanisms have demonstrated clinical efficacy in these regards, thus highlighting the utility of sequential ROS1 TKI therapy. Selective ROS1 inhibitors have yet to be developed, and thus the specific adverse effects of ROS1 inhibition cannot be deconvoluted from the toxicity profiles of the available multikinase inhibitors. Herein, we discuss the non-malignant and malignant biology of ROS1, the diagnostic challenges that ROS1 fusions present and the strategies to target ROS1 fusion proteins in both treatment-naive and acquired-resistance settings.
Collapse
Affiliation(s)
- Alexander Drilon
- Early Drug Development and Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Chelsea Jenkins
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Sudarshan Iyer
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Adam Schoenfeld
- Early Drug Development and Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Clare Keddy
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Monika A Davare
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
260
|
Forsythe A, Zhang W, Phillip Strauss U, Fellous M, Korei M, Keating K. A systematic review and meta-analysis of neurotrophic tyrosine receptor kinase gene fusion frequencies in solid tumors. Ther Adv Med Oncol 2020; 12:1758835920975613. [PMID: 33425024 PMCID: PMC7758559 DOI: 10.1177/1758835920975613] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction The research objective was to systematically review evidence on neurotrophic tyrosine receptor kinase (NTRK) gene fusion frequency in solid tumors. Methods Using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a systematic literature review (SLR) was conducted of studies published from January 1987 to 2 January 2020. Selected studies were appraised for use in meta-analysis, with frequency reported as a point estimate with confidence intervals, to estimate NTRK gene fusion tumor incidence and prevalence. Results The SLR identified 222 studies from North America (n = 122), Europe (n = 33), Asia (n = 41), Brazil (n = 5), Australia (n = 2), and multi-continental (n = 19) reporting NTRK gene fusion frequencies across 101 histologies. Studies were prospective (n = 43) and retrospective (n = 179). Testing methods involved DNA (n = 93), RNA (n = 72), combined DNA/RNA (n = 48), protein [immunohistochemistry (IHC), n = 5], and unreported (n = 5). Sample sizes ranged from 1 to 66,871. Of the 222 studies, 107 were suitable for meta-analysis. Highest NTRK gene fusion frequencies were reported in rare cancers: infantile/congenital fibrosarcoma (90.56%, 95% CI 67.42-100.00), secretory breast cancer (92.87%, 95% CI 72.62-100.00), and congenital mesoblastic nephroma (21.52%, 95% CI 13.06-32.20). Lower frequencies were reported in non-small cell lung cancer (0.17%, 95% CI 0.09-0.25), colorectal adenocarcinoma (0.26%, 95% CI 0.15-0.36), cutaneous melanoma (0.31%, 95% CI 0.07-0.55), and non-secretory breast carcinoma (0.60%, 95% CI 0.00-1.50). Reported frequency was ~0% for some cancers: mesothelioma, renal cell carcinoma, prostate cancer, and bone sarcoma. Estimated global overall NTRK gene fusion tumour incidence and 5-year prevalence in 2018 was 0.52 and 1.52 per 100,000 persons, respectively. Conclusion This research confirms the rarity and varying frequency of NTRK gene fusion across tumor types. Limitations included relatively low historic NTRK gene fusion testing and reporting, limited study samples for some cancers, and suboptimal molecular testing methods. In this rapidly developing area, gold-standard testing methods and companion diagnostics are needed to capture all NTRK gene fusions.
Collapse
Affiliation(s)
- Anna Forsythe
- Purple Squirrel Economics, 4 Lexington Ave, Suite 15K, New York, NY 10010, USA
| | - Wei Zhang
- Bayer Pharmaceuticals, Inc., Whippany, NJ, USA
| | | | | | | | | |
Collapse
|
261
|
Abstract
PURPOSE OF REVIEW Genetic aberrations resulting in tropomyosin receptor kinase (TRK) fusion proteins can drive oncogenesis and are postulated to occur in up to 1% of solid tumours. However, TRK fusions in adult sarcomas are rare and there is a significant challenge in identifying patients with sarcomas harbouring TRK fusions in the clinical setting. Despite a recent European Society of Medical Oncology consensus article regarding screening of tumours for TRK fusions, economical and practical limitations present a barrier to widespread screening of sarcomas. RECENT FINDINGS Larotrectinib and entrectinib are pan-TRK inhibitors which have both received FDA approval for the management of solid tumours harbouring NTRK fusions. Initial results of a number of clinical trials have demonstrated promising efficacy and safety data, including dramatic and durable responses in patients with sarcomas. As such, TRK inhibitors represent a promising treatment option in a small cohort of adult sarcoma patients, where currently treatment options are limited. The emergence of acquired resistance is a concern associated with TRK inhibitor therapy and a number of second-generation agents targeting TRK kinase mutations driving acquired resistance have entered early-phase clinical trials. SUMMARY With the growing appreciation of the implications of TRK fusions, this review will summarize the emerging clinical trial data of TRK inhibitors in sarcomas. Although in their infancy, clinical trial results are encouraging, and as further results and analyses are released, we will have a greater understanding of their impact on clinical practice and the management of patients with sarcomas.
Collapse
|
262
|
Thakral D, Gupta R, Sahoo RK, Verma P, Kumar I, Vashishtha S. Real-Time Molecular Monitoring in Acute Myeloid Leukemia With Circulating Tumor DNA. Front Cell Dev Biol 2020; 8:604391. [PMID: 33363162 PMCID: PMC7759522 DOI: 10.3389/fcell.2020.604391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
The clonal evolution of acute myeloid leukemia (AML), an oligoclonal hematological malignancy, is driven by a plethora of cytogenetic abnormalities, gene mutations, abnormal epigenetic patterns, and aberrant gene expressions. These alterations in the leukemic blasts promote clinically diverse manifestations with common characteristics of high relapse and drug resistance. Defining and real-time monitoring of a personalized panel of these predictive genetic biomarkers is rapidly being adapted in clinical setting for diagnostic, prognostic, and therapeutic decision-making in AML. A major challenge remains the frequency of invasive biopsy procedures that can be routinely performed for monitoring of AML disease progression. Moreover, a single-site biopsy is not representative of the tumor heterogeneity as it is spatially and temporally constrained and necessitates the understanding of longitudinal and spatial subclonal dynamics in AML. Hematopoietic cells are a major contributor to plasma cell-free DNA, which also contain leukemia-specific aberrations as the circulating tumor-derived DNA (ctDNA) fraction. Plasma cell-free DNA analysis holds immense potential as a minimally invasive tool for genomic profiling at diagnosis as well as clonal evolution during AML disease progression. With the technological advances and increasing sensitivity for detection of ctDNA, both genetic and epigenetic aberrations can be qualitatively and quantitatively evaluated. However, challenges remain in validating the utility of liquid biopsy tools in clinics, and universal recommendations are still awaited towards reliable diagnostics and prognostics. Here, we provide an overview on the scope of ctDNA analyses for prognosis, assessment of response to treatment and measurable residual disease, prediction of disease relapse, development of acquired resistance and beyond in AML.
Collapse
Affiliation(s)
- Deepshi Thakral
- Laboratory Oncology Unit, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Pramod Verma
- Laboratory Oncology Unit, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Indresh Kumar
- Laboratory Oncology Unit, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Sangeeta Vashishtha
- Laboratory Oncology Unit, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
263
|
Imyanitov EN, Iyevleva AG, Levchenko EV. Molecular testing and targeted therapy for non-small cell lung cancer: Current status and perspectives. Crit Rev Oncol Hematol 2020; 157:103194. [PMID: 33316418 DOI: 10.1016/j.critrevonc.2020.103194] [Citation(s) in RCA: 292] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Molecular testing has become a mandatory component of the non-small cell lung cancer (NSCLC) management. The detection of EGFR, BRAF and MET mutations as well as the analysis of ALK, ROS1, RET and NTRK translocations have already been incorporated in the NSCLC diagnostic standards, and the inhibitors of these kinases are in routine clinical use. There are emerging biomarkers, e.g., KRAS G12C substitutions and HER2 activating alterations, which are likely to enter NSCLC guidelines upon the approval of the corresponding drugs. In addition to genetic examination, NSCLCs are usually subjected to the analysis of PD-L1 protein expression in order to direct the use of immune checkpoint inhibitors. Comprehensive NSCLC testing for multiple predictive markers requires the analysis of distinct biological molecules (DNA, RNA, proteins) and, therefore, the involvement of different analytical platforms (PCR, DNA sequencing, immunohistochemistry, FISH). There are ongoing efforts aimed at the integration of multiple NSCLC molecular assays into a single diagnostic pipeline.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia; Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 195067, Russia.
| | - Aglaya G Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia
| | - Evgeny V Levchenko
- Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 195067, Russia; Department of Thoracic Oncology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia
| |
Collapse
|
264
|
Aguado C, Teixido C, Román R, Reyes R, Giménez-Capitán A, Marin E, Cabrera C, Viñolas N, Castillo S, Muñoz S, Arcocha A, López-Vilaró L, Sullivan I, Aldeguer E, Rodríguez S, Moya I, Viteri S, Cardona AF, Palmero R, Sainz C, Mesa-Guzmán M, Lozano MD, Aguilar-Hernández A, Martínez-Bueno A, González-Cao M, Gonzalvo E, Leenders WPJ, Rosell R, Montuenga LM, Prat A, Molina-Vila MA, Reguart N. Multiplex RNA-based detection of clinically relevant MET alterations in advanced non-small cell lung cancer. Mol Oncol 2020; 15:350-363. [PMID: 33236532 PMCID: PMC7858100 DOI: 10.1002/1878-0261.12861] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/23/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
MET inhibitors have shown activity in non‐small‐cell lung cancer patients (NSCLC) with MET amplification and exon 14 skipping (METΔex14). However, patient stratification is imperfect, and thus, response rates have varied widely. Here, we studied MET alterations in 474 advanced NSCLC patients by nCounter, an RNA‐based technique, together with next‐generation sequencing (NGS), fluorescence in situ hybridization (FISH), immunohistochemistry (IHC), and reverse transcriptase polymerase chain reaction (RT–PCR), exploring correlation with clinical benefit. Of the 474 samples analyzed, 422 (89%) yielded valid results by nCounter, which identified 13 patients (3%) with METΔex14 and 15 patients (3.5%) with very‐high MET mRNA expression. These two subgroups were mutually exclusive, displayed distinct phenotypes and did not generally coexist with other drivers. For METΔex14, 3/8 (37.5%) samples positive by nCounter tested negative by NGS. Regarding patients with very‐high MET mRNA, 92% had MET amplification by FISH and/or NGS. However, FISH failed to identify three patients (30%) with very‐high MET RNA expression, among which one received MET tyrosine kinase inhibitor treatment deriving clinical benefit. Our results indicate that quantitative mRNA‐based techniques can improve the selection of patients for MET‐targeted therapies.
Collapse
Affiliation(s)
- Cristina Aguado
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Cristina Teixido
- Thoracic Oncology Unit, Department of Pathology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ruth Román
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Roxana Reyes
- Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Ana Giménez-Capitán
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Elba Marin
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Carlos Cabrera
- Dr Rosell Oncology Institute, Dexeus University Hospital Quiron Salud Group, Barcelona, Spain
| | - Nuria Viñolas
- Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Sergi Castillo
- Division of Medical Oncology, Hospital General de Granollers, Barcelona, Spain
| | - Silvia Muñoz
- Division of Medical Oncology, Hospital General de Granollers, Barcelona, Spain
| | - Ainara Arcocha
- Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Laura López-Vilaró
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ivana Sullivan
- Division of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Erika Aldeguer
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Sonia Rodríguez
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Irene Moya
- Dr Rosell Oncology Institute, Dexeus University Hospital Quiron Salud Group, Barcelona, Spain
| | - Santiago Viteri
- Dr Rosell Oncology Institute, Dexeus University Hospital Quiron Salud Group, Barcelona, Spain.,Dr Rosell Oncology Institute, Teknon Medical Center, Quiron Salud Group, Barcelona, Spain
| | - Andrés Felipe Cardona
- Foundation for Clinical and Applied Cancer Research-FICMAC, Bogotá, Colombia.,Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
| | - Ramon Palmero
- Division of Medical Oncology, Catalan Institute of Oncology, L'Hospitalet, Barcelona, Spain
| | - Cristina Sainz
- Center for Applied Medical Research (CIMA), University of Navarra, Spain.,CIBERONC, Madrid, Spain
| | | | - Maria D Lozano
- CIBERONC, Madrid, Spain.,IDISNA, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | | | | | - María González-Cao
- Dr Rosell Oncology Institute, Dexeus University Hospital Quiron Salud Group, Barcelona, Spain
| | - Elena Gonzalvo
- Thoracic Oncology Unit, Department of Pathology, Hospital Clínic, Barcelona, Spain
| | - William P J Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Rafael Rosell
- Dr Rosell Oncology Institute, Dexeus University Hospital Quiron Salud Group, Barcelona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Luis M Montuenga
- Center for Applied Medical Research (CIMA), University of Navarra, Spain.,CIBERONC, Madrid, Spain.,IDISNA, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Miguel A Molina-Vila
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Noemi Reguart
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
265
|
Gupta S, Vanderbilt CM, Lin YT, Benhamida JK, Jungbluth AA, Rana S, Momeni-Boroujeni A, Chang JC, Mcfarlane T, Salazar P, Mullaney K, Middha S, Zehir A, Gopalan A, Bale TA, Ganly I, Arcila ME, Benayed R, Berger MF, Ladanyi M, Dogan S. A Pan-Cancer Study of Somatic TERT Promoter Mutations and Amplification in 30,773 Tumors Profiled by Clinical Genomic Sequencing. J Mol Diagn 2020; 23:253-263. [PMID: 33285287 DOI: 10.1016/j.jmoldx.2020.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/15/2020] [Accepted: 11/10/2020] [Indexed: 01/20/2023] Open
Abstract
TERT gene promoter mutations are known in multiple cancer types. Other TERT alterations remain poorly characterized. Sequencing data from 30,773 tumors analyzed by a hybridization capture next-generation sequencing assay (Memorial Sloan Kettering Cancer Center Integrated Mutation Profiling of Actionable Cancer Targets) were analyzed for the presence of TERT alterations. Promoter rearrangements (500 bases upstream of the transcriptional start site), hypermethylation (n = 57), and gene expression (n = 155) were evaluated for a subset of cases. Mutually exclusive and recurrent promoter mutations were identified at three hot spots upstream of the transcriptional start site in 11.3% of cases (-124: 74%; -146: 24%; and -138: <2%). Mutually exclusive amplification events were identified in another 2.3% of cases, whereas mutually exclusive rearrangements proximal to the TERT gene were seen in 24 cases. The highest incidence of TERT promoter mutations was seen in cutaneous melanoma (82%), whereas amplification events significantly outnumbered promoter mutations in well-differentiated/dedifferentiated liposarcoma (14.1% versus 2.4%) and adrenocortical carcinoma (13.6% versus 4.5%). Gene expression analysis suggests that the highest levels of gene expression are seen in cases with amplifications and rearrangements. Hypermethylation events upstream of the TERT coding sequence were not mutually exclusive with known pathogenic alterations. Studies aimed at defining the prevalence and prognostic impact of TERT alterations should incorporate other pathogenic TERT alterations as these may impact telomerase function.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yun-Te Lin
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jamal K Benhamida
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Satshil Rana
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tiffany Mcfarlane
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paulo Salazar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kerry Mullaney
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sumit Middha
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tejus A Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
266
|
Matter MS, Chijioke O, Savic S, Bubendorf L. Narrative review of molecular pathways of kinase fusions and diagnostic approaches for their detection in non-small cell lung carcinomas. Transl Lung Cancer Res 2020; 9:2645-2655. [PMID: 33489824 PMCID: PMC7815372 DOI: 10.21037/tlcr-20-676] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Abstract
The discovery of actionable oncogenic driver alterations has significantly improved treatment options for patients with advanced non-small cell lung cancer (NSCLC). In lung adenocarcinoma (LUAD), approved drugs or drugs in clinical development can target more than half of these altered oncogenic driver genes. In particular, several gene fusions have been discovered in LUAD, including ALK, ROS1, NTRK, RET, NRG1 and FGFR. All these fusions involve tyrosine kinases (TK), which are activated due to structural rearrangements on the DNA level. Although the overall prevalence of these fusions in LUAD is rare, their detection is extremely important, as they are linked to an excellent response to TK inhibitors. Therefore, reliable screening methods applicable to small tumor samples (biopsies and cytology specimens) are required in the diagnostic workup of advanced NSCLC. Several methods are at disposal in a routine laboratory to demonstrate, directly or indirectly, the presence of a gene fusion. These methods include immunohistochemistry (IHC), fluorescence in-situ hybridization (FISH), reverse transcriptase-polymerase chain reaction (RT-PCR), multiplex digital color-coded barcode technology or next-generation sequencing (NGS) either on DNA or RNA level. In our review, we will summarize the increasing number of relevant fusion genes in NSCLC, point out their underlining molecular mechanisms and discuss different methods for the detection of fusion genes.
Collapse
Affiliation(s)
| | - Obinna Chijioke
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Spasenija Savic
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Lukas Bubendorf
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
267
|
Dagogo-Jack I, Ritterhouse LL. The role of plasma genotyping in ALK- and ROS1-rearranged lung cancer. Transl Lung Cancer Res 2020; 9:2557-2570. [PMID: 33489818 PMCID: PMC7815348 DOI: 10.21037/tlcr-2019-cnsclc-09] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/11/2020] [Indexed: 01/04/2023]
Abstract
Several subsets of non-small cell lung cancer (NSCLC) are defined by the presence of oncogenic rearrangements that result in constitutive activation of a chimeric fusion protein. In NSCLCs that harbor ALK or ROS1 rearrangements, aberrant signaling from these fusion proteins can be overcome by potent and selective tyrosine kinase inhibitors (TKIs). These targeted therapies can induce durable responses and significantly improve prognostic outcomes. Historically, analysis of tissue biopsies was the primary approach to identifying key activating rearrangements. In recent years, non-invasive genotyping of tumor-derived nucleic acids in the circulation has gained ground as a strategy for determining the genetic composition of NSCLCs at diagnosis and throughout the disease course based on prospective and retrospective studies validating the utility of plasma analysis in heterogeneous populations of patients with metastatic NSCLC. Notably, these practice-changing studies predominantly included patients with NSCLCs with oncogenic mutations. Compared to other types of molecular alterations such as mutations and insertions/deletions, oncogenic rearrangements are more complex as they incorporate a variety of fusion partners and diverse breakpoints. Because of this structural complexity, detecting oncogenic rearrangements with plasma assays is more challenging than identifying disease-defining point mutations. In this review, we discuss technical aspects of plasma genotyping strategies and summarize findings from studies exploring plasma genotyping (including ctDNA analysis and profiling of nucleic acids contained in other plasma components) in two rearrangement-driven NSCLC subsets (ALK-rearranged and ROS1-rearranged).
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Lauren L. Ritterhouse
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
268
|
Xiong L, Li X, Chen D, Li S, Luo L. GPC1-ALK: A novel ALK fusion in a patient with pulmonary sarcomatoid carcinoma. Lung Cancer 2020; 151:104-105. [PMID: 33277054 DOI: 10.1016/j.lungcan.2020.11.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/22/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Lei Xiong
- Department of Cardiothoracic Surgery, The Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongsheng Chen
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
| | - Si Li
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
| | - Liguo Luo
- Department of Cardiothoracic Surgery, School of Medicine, Nanjing University, Nanjing, China.
| |
Collapse
|
269
|
Li W, Guo L, Liu Y, Dong L, Yang L, Chen L, Liu K, Shao Y, Ying J. Potential Unreliability of Uncommon ALK, ROS1, and RET Genomic Breakpoints in Predicting the Efficacy of Targeted Therapy in NSCLC. J Thorac Oncol 2020; 16:404-418. [PMID: 33248323 DOI: 10.1016/j.jtho.2020.10.156] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Variable genomic breakpoints have been identified through the application of target-capture DNA next-generation sequencing (NGS) for ALK, ROS1, and RET fusion detection in NSCLC. We investigated whether ALK, ROS1, and RET genomic breakpoint location can predict matched targeted therapy efficacy. METHODS NSCLCs were analyzed by DNA NGS, target-specific RNA NGS, whole-transcriptome sequencing, and immunohistochemistry. RESULTS In total, 3787 NSCLC samples were analyzed. DNA NGS detected ALK, ROS1, and RET fusions in 241, 59, and 76 cases, respectively. These fusions were divided into canonical (single EML4-ALK, CD74/EZR/TPM3/SDC4-ROS1, and KIF5B/CCDC6-RET fusions), noncanonical (single non-EML4-ALK, non-CD74/EZR/TPM3/SDC4-ROS1, and non-KIF5B/CCDC6-RET fusions), and primary/reciprocal (both primary and reciprocal rearrangements were detected) subtypes on the basis of genomic breakpoint position, and noncanonical and primary/reciprocal subtypes were defined as uncommon fusions. Further RNA sequencing and immunohistochemistry revealed that six of 47 (12.8%) uncommon fusions were actually nonproductive rearrangements that generated no aberrant transcripts or proteins. Moreover, genomic breakpoints of canonical ALK and RET, but not ROS1, fusions always predicted breakpoints at the transcript level, whereas 85.4% (35 of 41) of uncommon fusions actually produced canonical fusion transcripts. Patients with uncommon ALK fusion (n = 31) who received first-line crizotinib exhibited shorter median progression-free survival than those with canonical ALK fusion (n = 53, 8.4 mo versus 12.0 mo, p = 0.004). However, no difference in progression-free survival was observed when only ALK RNA or protein-positive cases were analyzed (p = 0.185). CONCLUSIONS Uncommon ALK, ROS1, and RET genomic breakpoint is an unreliable predictor of matched targeted therapy efficacy. Functional validation by RNA or protein assay may add value for the accurate detection and interpretation of rare fusions.
Collapse
Affiliation(s)
- Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Li Chen
- HeliTec Biotechnologies, Shenzhen, Guangdong, People's Republic of China
| | - Kaihua Liu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, People's Republic of China
| | - Yang Shao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, People's Republic of China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
270
|
Ogura K, Somwar R, Hmeljak J, Magnan H, Benayed R, Momeni Boroujeni A, Bowman AS, Mattar MS, Khodos I, de Stanchina E, Jungbluth A, Asher M, Odintsov I, Hartono AB, LaQuaglia MP, Slotkin E, Pratilas CA, Lee SB, Spraggon L, Ladanyi M. Therapeutic Potential of NTRK3 Inhibition in Desmoplastic Small Round Cell Tumor. Clin Cancer Res 2020; 27:1184-1194. [PMID: 33229458 DOI: 10.1158/1078-0432.ccr-20-2585] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/27/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Desmoplastic small round cell tumor (DSRCT) is a highly lethal intra-abdominal sarcoma of adolescents and young adults. DSRCT harbors a t(11;22)(p13:q12) that generates the EWSR1-WT1 chimeric transcription factor, the key oncogenic driver of DSRCT. EWSR1-WT1 rewires global gene expression networks and activates aberrant expression of targets that together mediate oncogenesis. EWSR1-WT1 also activates a neural gene expression program. EXPERIMENTAL DESIGN Among these neural markers, we found prominent expression of neurotrophic tyrosine kinase receptor 3 (NTRK3), a druggable receptor tyrosine kinase. We investigated the regulation of NTRK3 by EWSR1-WT1 and its potential as a therapeutic target in vitro and in vivo, the latter using novel patient-derived models of DSRCT. RESULTS We found that EWSR1-WT1 binds upstream of NTRK3 and activates its transcription. NTRK3 mRNA is highly expressed in DSRCT compared with other major chimeric transcription factor-driven sarcomas and most DSRCTs are strongly immunoreactive for NTRK3 protein. Remarkably, expression of NTRK3 kinase domain mRNA in DSRCT is also higher than in cancers with NTRK3 fusions. Abrogation of NTRK3 expression by RNAi silencing reduces growth of DSRCT cells and pharmacologic targeting of NTRK3 with entrectinib is effective in both in vitro and in vivo models of DSRCT. CONCLUSIONS Our results indicate that EWSR1-WT1 directly activates NTRK3 expression in DSRCT cells, which are dependent on its expression and activity for growth. Pharmacologic inhibition of NTRK3 by entrectinib significantly reduces growth of DSRCT cells both in vitro and in vivo, providing a rationale for clinical evaluation of NTRK3 as a therapeutic target in DSRCT.
Collapse
Affiliation(s)
- Koichi Ogura
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Romel Somwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julija Hmeljak
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heather Magnan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Anita S Bowman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marissa S Mattar
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Inna Khodos
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Achim Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marina Asher
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Igor Odintsov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alifiani B Hartono
- Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Michael P LaQuaglia
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Sean Bong Lee
- Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Lee Spraggon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
271
|
Guo R, Offin M, Brannon AR, Chang J, Chow A, Delasos L, Girshman J, Wilkins O, McCarthy CG, Makhnin A, Falcon C, Scott K, Tian Y, Cecchi F, Hembrough T, Alex D, Shen R, Benayed R, Li BT, Rudin CM, Kris MG, Arcila ME, Rekhtman N, Paik P, Zehir A, Drilon A. MET Exon 14-altered Lung Cancers and MET Inhibitor Resistance. Clin Cancer Res 2020; 27:799-806. [PMID: 33172896 DOI: 10.1158/1078-0432.ccr-20-2861] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/17/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE MET tyrosine kinase inhibitors (TKIs) can achieve modest clinical outcomes in MET exon 14-altered lung cancers, likely secondary to primary resistance. Mechanisms of primary resistance remain poorly characterized and comprehensive proteomic analyses have not previously been performed. EXPERIMENTAL DESIGN We performed hybrid capture-based DNA sequencing, targeted RNA sequencing, cell-free DNA sequencing, selected reaction monitoring mass spectrometry (SRM-MS), and immunohistochemistry on patient samples of MET exon 14-altered lung cancers treated with a MET TKI. Associations between overall response rate (ORR), progression-free survival (PFS), and putative genomic alterations and MET protein expression were evaluated. RESULTS Seventy-five of 168 MET exon 14-altered lung cancers received a MET TKI. Previously undescribed (zygosity, clonality, whole-genome duplication) and known (copy-number focality, tumor mutational burden, mutation region/type) genomic factors were not associated with ORR/PFS (P > 0.05). In contrast, MET expression was associated with MET TKI benefit. Only cases with detectable MET expression by SRM-MS (N = 15) or immunochemistry (N = 22) responded to MET TKI therapy, and cancers with H-score ≥ 200 had a higher PFS than cancers below this cutoff (10.4 vs. 5.5 months, respectively; HR, 3.87; P = 0.02). CONCLUSIONS In MET exon 14-altered cancers treated with a MET TKI, a comprehensive analysis of previously unknown and known genomic factors did not identify a genomic mechanism of primary resistance. Instead, MET expression correlated with benefit, suggesting the potential role of interrogating the proteome in addition to the genome in confirmatory prospective trials.
Collapse
Affiliation(s)
- Robin Guo
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Offin
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - A Rose Brannon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew Chow
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas Delasos
- Department of Medicine, UConn Health, Farmington, Connecticut
| | - Jeffrey Girshman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Olivia Wilkins
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Caroline G McCarthy
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alex Makhnin
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina Falcon
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | - Deepu Alex
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronglai Shen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bob T Li
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Charles M Rudin
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark G Kris
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul Paik
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
272
|
D’Angelo A, Sobhani N, Chapman R, Bagby S, Bortoletti C, Traversini M, Ferrari K, Voltolini L, Darlow J, Roviello G. Focus on ROS1-Positive Non-Small Cell Lung Cancer (NSCLC): Crizotinib, Resistance Mechanisms and the Newer Generation of Targeted Therapies. Cancers (Basel) 2020; 12:3293. [PMID: 33172113 PMCID: PMC7694780 DOI: 10.3390/cancers12113293] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/18/2022] Open
Abstract
The treatment of patients affected by non-small cell lung cancer (NSCLC) has been revolutionised by the discovery of druggable mutations. ROS1 (c-ros oncogene) is one gene with druggable mutations in NSCLC. ROS1 is currently targeted by several specific tyrosine kinase inhibitors (TKIs), but only two of these, crizotinib and entrectinib, have received Food and Drug Administration (FDA) approval. Crizotinib is a low molecular weight, orally available TKI that inhibits ROS1, MET and ALK and is considered the gold standard first-line treatment with demonstrated significant activity for lung cancers harbouring ROS1 gene rearrangements. However, crizotinib resistance often occurs, making the treatment of ROS1-positive lung cancers more challenging. A great effort has been undertaken to identify a new generation or ROS1 inhibitors. In this review, we briefly introduce the biology and role of ROS1 in lung cancer and discuss the underlying acquired mechanisms of resistance to crizotinib and the promising new agents able to overcome resistance mechanisms and offer alternative efficient therapies.
Collapse
Affiliation(s)
- Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Robert Chapman
- University College London Hospitals NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK;
| | - Stefan Bagby
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Carlotta Bortoletti
- Department of Dermatology, University of Padova, via Vincenzo Gallucci 4, 35121 Padova, Italy;
| | - Mirko Traversini
- Unità Operativa Anatomia Patologica, Ospedale Maggiore Carlo Alberto Pizzardi, AUSL Bologna, Largo Bartolo Nigrisoli 2, 40100 Bologna, Italy;
| | - Katia Ferrari
- Respiratory Medicine, Careggi University Hospital, 50139 Florence, Italy;
| | - Luca Voltolini
- Thoracic Surgery Unit, Careggi University Hospital, Largo Brambilla, 1, 50134 Florence, Italy;
| | - Jacob Darlow
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy;
| |
Collapse
|
273
|
Marmarelis ME, Chiarle R, Ambrogio C, Aggarwal C. A LIBRETTO to orchestrate targeted therapy. NATURE CANCER 2020; 1:1038-1040. [PMID: 35122069 DOI: 10.1038/s43018-020-00144-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Melina E Marmarelis
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roberto Chiarle
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| | - Charu Aggarwal
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
274
|
Sholl LM, Hwang DH. Expanding the utility of cytology preparations in cancer biomarker testing. Cancer Cytopathol 2020; 129:337-340. [PMID: 33119210 DOI: 10.1002/cncy.22380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - David H Hwang
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
275
|
Conde E, Hernandez S, Sanchez E, Regojo RM, Camacho C, Alonso M, Martinez R, Lopez-Rios F. Pan-TRK Immunohistochemistry: An Example-Based Practical Approach to Efficiently Identify Patients With NTRK Fusion Cancer. Arch Pathol Lab Med 2020; 145:1031-1040. [PMID: 33112951 DOI: 10.5858/arpa.2020-0400-ra] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Food and Drug Administration-approved TRK inhibitors with impressive overall response rates are now available for patients with multiple cancer types that harbor NTRK rearrangements, yet the identification of NTRK fusions remains a difficult challenge. These alterations are highly recurrent in extremely rare malignancies or can be detected in exceedingly small subsets of common tumor types. A 2-step approach has been proposed, involving a screening by immunohistochemistry (IHC) followed by a confirmatory method (fluorescence in situ hybridization, reverse transcriptase-polymerase chain reaction, or next-generation sequencing) in cases expressing the protein. However, there is no interpretation guide for any of the available IHC clones. OBJECTIVE.— To provide a pragmatic update on the use of pan-TRK IHC. Selected examples of the different IHC staining patterns across multiple histologies are shown. DATA SOURCES.— Primary literature review with PubMed, combined with personal diagnostic and research experience. CONCLUSIONS.— In-depth knowledge of pan-TRK IHC will help pathologists implement a rational approach to the detection of NTRK fusions in human malignancies.
Collapse
Affiliation(s)
- Esther Conde
- From Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HMHospitales, CIBERONC, Madrid, Spain (Conde, Lopez-Rios)
| | - Susana Hernandez
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HMHospitales, Madrid, Spain (Hernandez, Sanchez, Alonso, Martinez)
| | - Elena Sanchez
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HMHospitales, Madrid, Spain (Hernandez, Sanchez, Alonso, Martinez)
| | | | - Carmen Camacho
- Pathology, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain (Camacho). Conde and Hernandez contributed equally as co-first authors
| | - Marta Alonso
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HMHospitales, Madrid, Spain (Hernandez, Sanchez, Alonso, Martinez)
| | - Rebeca Martinez
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HMHospitales, Madrid, Spain (Hernandez, Sanchez, Alonso, Martinez)
| | - Fernando Lopez-Rios
- From Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HMHospitales, CIBERONC, Madrid, Spain (Conde, Lopez-Rios)
| |
Collapse
|
276
|
Dagogo-Jack I, Lennerz JK. Personalized Diagnostic Workflows: The Next Wave of Precision Medicine in NSCLC. J Thorac Oncol 2020; 15:888-890. [PMID: 32471563 DOI: 10.1016/j.jtho.2020.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
277
|
Lamberti G, Andrini E, Sisi M, Rizzo A, Parisi C, Di Federico A, Gelsomino F, Ardizzoni A. Beyond EGFR, ALK and ROS1: Current evidence and future perspectives on newly targetable oncogenic drivers in lung adenocarcinoma. Crit Rev Oncol Hematol 2020; 156:103119. [PMID: 33053439 DOI: 10.1016/j.critrevonc.2020.103119] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide. In the past decade EGFR, ALK and ROS1 TKIs lead to an unprecedented survival improvement of oncogene-addicted NSCLC patients, with better toxicity profile compared to chemotherapy. In recent years the implementation of high-throughput sequencing platforms led to the identification of uncommon molecular alterations in oncogenic drivers, such as BRAF, MET, RET, HER2 and NTRK. Moreover, newly developed drugs have been found to be active against hard to target drivers, such as KRAS. Specific TKIs targeting these genomic alterations are currently in clinical development and showed impressive activity and survival improvement, leading to FDA-accelerated approval for some of them. However, virtually all patients develop resistance to TKIs by on-target or off-target mechanisms. Here we review the clinicopathological features, the emerging targeted therapies and mechanisms of resistance and strategies to overcome them of KRAS, BRAF, MET, RET, HER2 and NTRK-addicted advanced NSCLCs.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Elisa Andrini
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Monia Sisi
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessandro Rizzo
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Claudia Parisi
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessandro Di Federico
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Francesco Gelsomino
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy.
| | - Andrea Ardizzoni
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy; Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| |
Collapse
|
278
|
Arcila ME, Yang SR, Momeni A, Mata DA, Salazar P, Chan R, Elezovic D, Benayed R, Zehir A, Buonocore DJ, Rekhtman N, Lin O, Ladanyi M, Nafa K. Ultrarapid EGFR Mutation Screening Followed by Comprehensive Next-Generation Sequencing: A Feasible, Informative Approach for Lung Carcinoma Cytology Specimens With a High Success Rate. JTO Clin Res Rep 2020; 1:100077. [PMID: 33511359 PMCID: PMC7839984 DOI: 10.1016/j.jtocrr.2020.100077] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION For patients with advanced NSCLC, cytologic samples may be the only diagnostic specimen available for molecular profiling. Although both rapid and comprehensive assessment are essential in this setting, an integrated multitest approach remains an important strategy in many laboratories, despite the risks and challenges when working with scant samples. In this study, we describe our experience and high success rate in using a multitest approach, focusing on the clinical validation and incorporation of ultrarapid EGFR testing using the Idylla system followed by comprehensive next-generation sequencing (NGS). METHODS Cytology samples received for routine molecular testing were included in this study. The performance characteristics of the EGFR Idylla assay were assessed; tissue suitability parameters and interpretation criteria to supplement automated mutation calling were established. The assay performance was monitored for 1 year, comparing the results with those of concurrent NGS testing by MSK-IMPACT (primarily) or MSK-AmpliSeq and MSK-Fusion solid panel in a subset of cases. RESULTS Overall, 301 samples were studied; 83 samples were included in validation (60.2% [50 of 83] were positive for EGFR mutations). Concordance with the reference method was 96.4% (80 of 83) of the samples with excellent reproducibility. The limit of detection was variable depending on the total tissue input and the specific mutation tested. Unextracted tissue inputs that maintained total EGFR cycle of quantification at less than 23 allowed all mutations to be detected if present at greater than 5% variant allele frequency. Mutations could be detected at 1% variant allele frequency with total EGFR cycle of quantification of 18. During the clinical implementation phase, 218 NSCLC samples were tested by Idylla (24.3% [53 of 218] were EGFR mutation positive). Concurrent NGS testing was requested on 165 samples and successfully performed on 96.4% (159 of 165) of the samples. The Idylla automated results were concordant with those obtained by NGS in 96.2% (153 of 159) of cases and improved to 98.7% (157 of 159) after incorporation of manual review criteria to supplement automated calling, resulting in a diagnostic sensitivity of 95.6% (95% confidence interval: 84.9%-99.5%). In general, 9% (14 of 159) of the cases tested by NGS had EGFR mutations not covered by the Idylla assay, primarily insertions in exon 19 and 20 and minor mutations cooccurring with canonical sensitizing mutations. CONCLUSIONS Comprehensive molecular testing is feasible and has a high success rate in NSCLC cytology samples when using a multitest approach. Testing with the Idylla system enables rapid and accurate determination of the EGFR status without compromising subsequent NGS testing.
Collapse
Affiliation(s)
- Maria E. Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Soo-Ryum Yang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amir Momeni
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Douglas A. Mata
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paulo Salazar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roger Chan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Elezovic
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Darren J. Buonocore
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Oscar Lin
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Khedoudja Nafa
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
279
|
PLEKHH2-ALK: A Novel In-frame Fusion With Durable Response to Alectinib: Utilizing RNA Sequencing in Search for Hidden Gene Fusions Susceptible to Targeted Therapy. Clin Lung Cancer 2020; 22:e51-e53. [PMID: 32893122 DOI: 10.1016/j.cllc.2020.07.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 11/23/2022]
|
280
|
Cooper AJ, Kobayashi Y, Kim D, Clifford SE, Kravets S, Dahlberg SE, Chambers ES, Li J, Rangachari D, Nguyen T, Costa DB, Rabin MS, Wagle N, Sholl LM, Jänne PA, Oxnard GR. Identification of a RAS-activating TMEM87A-RASGRF1 Fusion in an Exceptional Responder to Sunitinib with Non-Small Cell Lung Cancer. Clin Cancer Res 2020; 26:4072-4079. [PMID: 32312893 PMCID: PMC7415568 DOI: 10.1158/1078-0432.ccr-20-0397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/24/2020] [Accepted: 04/10/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We pursued genomic analysis of an exceptional responder with non-small cell lung cancer (NSCLC) through a multi-platform effort to discover novel oncogenic targets. EXPERIMENTAL DESIGN In this open-label, single-arm phase II study (NCT01829217), an enriched cohort of patients with advanced NSCLC was treated with the multi-kinase inhibitor sunitinib. The primary endpoint was objective response rate. Tissue was collected for multi-platform genomic analysis of responders, and a candidate oncogene was validated using in vitro models edited by CRISPR-Cas9. RESULTS Of 13 patients enrolled, 1 patient (8%), a never smoker, had a partial response lasting 33 months. Genomic analysis of the responder identified no oncogenic variant using multi-platform DNA analysis including hotspot allelotyping, massively parallel hybrid-capture next-generation sequencing, and whole-exome sequencing. However, bulk RNA-sequencing (RNA-seq) revealed a novel fusion, TMEM87A-RASGRF1, with high overexpression of the fusion partners. RASGRF1 encodes a guanine exchange factor which activates RAS from GDP-RAS to GTP-RAS. Oncogenicity was demonstrated in NIH/3T3 models with intrinsic TMEM87A-RASGRF1 fusion. In addition, activation of MAPK was shown in PC9 models edited to express this fusion, although sensitivity to MAPK inhibition was seen without apparent sensitivity to sunitinib. CONCLUSIONS Sunitinib exhibited limited activity in this enriched cohort of patients with advanced NSCLC. Nonetheless, we find that RNA-seq of exceptional responders represents a potentially underutilized opportunity to identify novel oncogenic targets including oncogenic activation of RASGRF1.
Collapse
Affiliation(s)
- Alissa J Cooper
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yoshihisa Kobayashi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dewey Kim
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Sarah E Clifford
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sasha Kravets
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Suzanne E Dahlberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Emily S Chambers
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jiaqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Deepa Rangachari
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center (a member of Beth Israel Lahey Health), Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Tom Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daniel B Costa
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center (a member of Beth Israel Lahey Health), Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Michael S Rabin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Nikhil Wagle
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Geoffrey R Oxnard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
281
|
Offin M, Luo J, Guo R, Lyo JK, Falcon C, Dienstag J, Wilkins O, Chang J, Rudin CM, Riely G, Rekhtman N, Arcila ME, Heller G, Ladanyi M, Li BT, Kris MG, Paik P, Drilon A. CNS Metastases in Patients With MET Exon 14-Altered Lung Cancers and Outcomes With Crizotinib. JCO Precis Oncol 2020; 4:2000098. [PMID: 32923895 DOI: 10.1200/po.20.00098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2020] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Although MET exon 14 (METex14)-altered lung cancers were first identified more than a decade and a half ago, the frequency of CNS metastatic disease remains poorly defined. Furthermore, the seminal trial of crizotinib in these patients (PROFILE 1001) did not report patterns of CNS response or progression. PATIENTS AND METHODS Patients with pathologically confirmed, advanced non-small-cell lung cancers (NSCLC) harboring a METex14 alteration by targeted DNA/RNA sequencing were studied. The incidence of brain metastases and the outcomes of MET inhibition with crizotinib were analyzed. RESULTS Eighty-three patients with METex14-altered metastatic NSCLC were identified. The incidence of CNS metastases at diagnosis was 17% (95% CI, 10% to 27%). The lifetime incidence was 36% (95% CI, 26% to 47%); 83% of patients had parenchymal disease, and 17% had leptomeningeal disease. The probability of having brain metastasis at 1, 2, and 3 years was 24%, 35%, and 38%, respectively. Fifty-four patients received crizotinib. The median time to radiologic CNS progression was 5.8 months (range, 3.7-20.0 months). Patterns of crizotinib progression were as follows: intracranial only in 10% of patients, intracranial and extracranial in 12%, and extracranial only in 78%. In patients with brain metastases before treatment, the median time on crizotinib was 7.5 months (range, 7.2-11.7 months). CONCLUSION CNS metastases, including leptomeningeal disease, occurred in more than a third of patients with METex14-altered lung cancers. In crizotinib-treated patients with or without CNS metastases, CNS failure was seen in less than a quarter of patients on progression.
Collapse
Affiliation(s)
- Michael Offin
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Jia Luo
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Robin Guo
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - John K Lyo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christina Falcon
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Jordan Dienstag
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Olivia Wilkins
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Jason Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charles M Rudin
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Gregory Riely
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Glenn Heller
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bob T Li
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Mark G Kris
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Paul Paik
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Alexander Drilon
- Thoracic Oncology and Early Drug Development Services, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| |
Collapse
|
282
|
Yang SR, Schultheis AM, Yu H, Mandelker D, Ladanyi M, Büttner R. Precision medicine in non-small cell lung cancer: Current applications and future directions. Semin Cancer Biol 2020; 84:184-198. [PMID: 32730814 DOI: 10.1016/j.semcancer.2020.07.009] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/24/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022]
Abstract
Advances in biomarkers, targeted therapies, and immuno-oncology have transformed the clinical management of patients with advanced NSCLC. For oncogene-driven tumors, there are highly effective targeted therapies against EGFR, ALK, ROS1, BRAF, TRK, RET, and MET. In addition, investigational therapies for KRAS, NRG1, and HER2 have shown promising results and may become standard-of-care in the near future. In parallel, immune-checkpoint therapy has emerged as an indispensable treatment modality, especially for patients lacking actionable oncogenic drivers. While PD-L1 expression has shown modest predictive utility, biomarkers for immune-checkpoint inhibition in NSCLC have remained elusive and represent an area of active investigation. Given the growing importance of biomarkers, optimal utilization of small tissue biopsies and alternative genotyping methods using circulating cell-free DNA have become increasingly integrated into clinical practice. In this review, we will summarize the current landscape and emerging trends in precision medicine for patients with advanced NSCLC with a special focus on predictive biomarker testing.
Collapse
Affiliation(s)
- Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | | | - Helena Yu
- Memorial Sloan Kettering Cancer Center, Department of Medicine, United States
| | - Diana Mandelker
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | - Reinhard Büttner
- University Hospital of Cologne, Department of Pathology, Germany.
| |
Collapse
|
283
|
Bruno R, Fontanini G. Next Generation Sequencing for Gene Fusion Analysis in Lung Cancer: A Literature Review. Diagnostics (Basel) 2020; 10:E521. [PMID: 32726941 PMCID: PMC7460167 DOI: 10.3390/diagnostics10080521] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023] Open
Abstract
Gene fusions have a pivotal role in non-small cell lung cancer (NSCLC) precision medicine. Several techniques can be used, from fluorescence in situ hybridization and immunohistochemistry to next generation sequencing (NGS). Although several NGS panels are available, gene fusion testing presents more technical challenges than other variants. This is a PubMed-based narrative review aiming to summarize NGS approaches for gene fusion analysis and their performance on NSCLC clinical samples. The analysis can be performed at DNA or RNA levels, using different target enrichment (hybrid-capture or amplicon-based) and sequencing chemistries, with both custom and commercially available panels. DNA sequencing evaluates different alteration types simultaneously, but large introns and repetitive sequences can impact on the performance and it does not discriminate between expressed and unexpressed gene fusions. RNA-based targeted approach analyses and quantifies directly fusion transcripts and is more accurate than DNA panels on tumor tissue, but it can be limited by RNA quality and quantity. On liquid biopsy, satisfying data have been published on circulating tumor DNA hybrid-capture panels. There is not a perfect method for gene fusion analysis, but NGS approaches, though still needing a complete standardization and optimization, present several advantages for the clinical practice.
Collapse
Affiliation(s)
- Rossella Bruno
- Unit of Pathological Anatomy, University Hospital of Pisa, Via Roma 67, 56126 Pisa, Italy;
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
| |
Collapse
|
284
|
Stinchcombe TE. Current management of RET rearranged non-small cell lung cancer. Ther Adv Med Oncol 2020; 12:1758835920928634. [PMID: 32782485 PMCID: PMC7385825 DOI: 10.1177/1758835920928634] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/01/2020] [Indexed: 12/14/2022] Open
Abstract
The identification of oncogenic drivers, and the subsequent development of targeted therapies established biomarker-based care for metastatic non-small cell lung cancer (NSCLC). Biomarker testing is standard of care in NSCLC patients with adenocarcinoma because multiple targeted therapies are available. Rearranged during transfection (RET) rearrangements were identified as oncogenic drivers in NSCLC, and are more common among younger patients, adenocarcinoma histology, and patients with a history of never smoking. The prevalence is estimated to be 1–2% among patients with adenocarcinoma histology. The most common rearrangement is between intron 11 of the RET gene and intron 15 of the KIF5B gene, and the next most frequent rearrangement is with the CCDC6 gene. RET rearrangements lead to constitutive activation of the RET tyrosine kinase and increased cell proliferation, migration, and survival. Phase II studies investigated the activity of multi-targeted tyrosine kinase inhibitors in patients with NSCLC with a confirmed RET rearrangement. These agents have limited potency against RET, and activity against the epidermal growth factor receptor and vascular endothelial growth factor pathways. These agents revealed modest activity, and were poorly tolerated due to the off-target toxicities. These struggles contributed to the development of more potent and specific RET tyrosine kinase inhibitors. Preliminary results from early phase trials of selpercatinib (LOXO-292) and pralsetinib (BLU-667) revealed promising efficacy and improved tolerability. The availability of these agents will make routine testing for RET rearrangements a priority.
Collapse
|
285
|
El Dika I, Bowman AS, Berger MF, Capanu M, Chou JF, Benayed R, Zehir A, Shia J, O'Reilly EM, Klimstra DS, Solit DB, Abou-Alfa GK. Molecular profiling and analysis of genetic aberrations aimed at identifying potential therapeutic targets in fibrolamellar carcinoma of the liver. Cancer 2020; 126:4126-4135. [PMID: 32663328 DOI: 10.1002/cncr.32960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/23/2020] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Fibrolamellar carcinoma (FLC) is a rare primary liver cancer of young adults. A functional chimeric transcript resulting from the in-frame fusion of the DNAJ homolog, subfamily B, member 1 (DNAJB1), and the catalytic subunit of protein kinase A (PRKACA) genes on chromosome 19 is believed to be unique in FLC, with a possible role in pathogenesis, yet with no established therapeutic value. The objective of the current study was to understand the molecular landscape of FLC and to identify potential novel therapeutic targets. METHODS Archival fresh, formalin-fixed, paraffin-embedded samples from patients with FLC who prospectively consented to an institutional review board-approved protocol were analyzed using Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT), a next-generation sequencing assay encompassing up to 468 key cancer genes. Custom targeted RNA-Seq was performed in selected patients. Demographics, treatment, and outcome data were collected prospectively. Survival outcomes were estimated and correlated with mutation and/or copy number alterations. RESULTS A total of 33 tumor samples from 31 patients with FLC were analyzed. The median age of the patients at the time of diagnosis was 18 years and approximately 53% were women. The DNAJB1-PRKACA fusion transcript was detected in 100% of patients. In 10 of 31 patients in which MSK-IMPACT did not detect the fusion, its presence was confirmed by targeted RNA-Seq. TERT promoter mutation was the second most common, and was detected in 7 patients. The median follow up was 30 months (range, 6-153 months). The 3-year overall survival rate was 84% (95% CI, 61%-93%). CONCLUSIONS The DNAJB1-PRKACA fusion transcript is nonspecific and nonsensitive to FLC. Its potential therapeutic value currently is under evaluation. Opportunities currently are under development for therapy that may be driven or related to the DNAJB1-PRKACA fusion transcript or any therapeutic target identified from next-generation sequencing in patients with FLC.
Collapse
Affiliation(s)
- Imane El Dika
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| | - Anita S Bowman
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael F Berger
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| | - Marinela Capanu
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joanne F Chou
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ryma Benayed
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ahmet Zehir
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jinru Shia
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| | - David S Klimstra
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| | - David B Solit
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| | - Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell College of Medicine, New York, New York, USA
| |
Collapse
|
286
|
Solomon JP, Benayed R, Hechtman JF, Ladanyi M. Identifying patients with NTRK fusion cancer. Ann Oncol 2020; 30 Suppl 8:viii16-viii22. [PMID: 31738428 PMCID: PMC6859817 DOI: 10.1093/annonc/mdz384] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Due to the efficacy of tropomyosin receptor kinase (TRK) inhibitor therapy and the recent Food and Drug Administration approval of larotrectinib, it is now clinically important to accurately and efficiently identify patients with neurotrophic TRK (NTRK) fusion-driven cancer. These oncogenic fusions occur when the kinase domain of NTRK1, NTRK2 or NTRK3 fuse with any of a number of N-terminal partners. NTRK fusions are characteristic of a few rare types of cancer, such as secretory carcinoma of the breast or salivary gland and infantile fibrosarcoma, but they are also infrequently seen in some common cancers, such as melanoma, glioma and carcinomas of the thyroid, lung and colon. There are multiple methods for identifying NTRK fusions, including pan-TRK immunohistochemistry, fluorescence in situ hybridisation and sequencing methods, and the advantages and drawbacks of each are reviewed here. While testing algorithms will obviously depend on availability of various testing modalities and economic considerations for each individual laboratory, we propose triaging specimens based on histology and other molecular findings to most efficiently identify tumours harbouring these treatable oncogenic fusions.
Collapse
Affiliation(s)
- J P Solomon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - R Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - M Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
287
|
The Evolving Landscape of Resistance to Osimertinib. J Thorac Oncol 2020; 15:18-21. [PMID: 31864549 DOI: 10.1016/j.jtho.2019.11.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022]
|
288
|
López-Ríos F, Paz-Ares L, Sanz J, Isla D, Pijuan L, Felip E, Gómez-Román JJ, de Castro J, Conde E, Garrido P. [Updated guidelines for predictive biomarker testing in advanced non-small-cell lung cancer: A National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2020; 53:167-181. [PMID: 32650968 DOI: 10.1016/j.patol.2019.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023]
Abstract
In 2011, the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathology (SEAP) initiated a joint project to establish guidelines for biomarker testing in patients with advanced non-small-cell lung cancer based on the information available at the time. As this field is constantly evolving, these guidelines were updated in 2012 and 2015 and now in 2019. Current evidence suggests it should be mandatory to test all patients with this kind of advanced lung cancer for EGFR and BRAF mutations, ALK and ROS1 rearrangements and PD-L1 expression. The growing need to study other emerging biomarkers has promoted the routine use of massive sequencing (next-generation sequencing, NGS). However, the coordination of every professional involved and the prioritisation of the most suitable tests and technologies for each case remain a challenge.
Collapse
Affiliation(s)
- Fernando López-Ríos
- Departamento de Patología-Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, CIBERONC, Madrid, España.
| | - Luis Paz-Ares
- Servicio de Oncología Médica, Hospital Universitario 12 de Octubre, Madrid, España
| | - Julián Sanz
- Departamento de Patología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, España
| | - Dolores Isla
- Servicio de Oncología Médica, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| | - Lara Pijuan
- Departamento de Patología, Hospital del Mar, Barcelona, España
| | - Enriqueta Felip
- Departamento de Oncología Médica, Hospital Universitari Vall d'Hebron, Barcelona, España
| | - José Javier Gómez-Román
- Departamento de Patología, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, IDIVAL, Santander, España
| | - Javier de Castro
- Departamento de Oncología Médica, Hospital Universitario La Paz, Madrid, España
| | - Esther Conde
- Departamento de Patología-Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, CIBERONC, Madrid, España
| | - Pilar Garrido
- Departamento de Oncología Médica, Hospital Universitario Ramón y Cajal, Universidad Alcalá, IRYCIS, CIBERONC, Madrid, España
| |
Collapse
|
289
|
Guo R, Luo J, Chang J, Rekhtman N, Arcila M, Drilon A. MET-dependent solid tumours - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol 2020; 17:569-587. [PMID: 32514147 DOI: 10.1038/s41571-020-0377-z] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Attempts to develop MET-targeted therapies have historically focused on MET-expressing cancers, with limited success. Thus, MET expression in the absence of a genomic marker of MET dependence is a poor predictor of benefit from MET-targeted therapy. However, owing to the development of more sensitive methods of detecting genomic alterations, high-level MET amplification and activating MET mutations or fusions are all now known to be drivers of oncogenesis. MET mutations include those affecting the kinase or extracellular domains and those that result in exon 14 skipping. The activity of MET tyrosine kinase inhibitors varies by MET alteration category. The likelihood of benefit from MET-targeted therapies increases with increasing levels of MET amplification, although no consensus exists on the optimal diagnostic cut-off point for MET copy number gains identified using fluorescence in situ hybridization and, in particular, next-generation sequencing. Several agents targeting exon 14 skipping alterations are currently in clinical development, with promising data available from early-phase trials. By contrast, the therapeutic implications of MET fusions remain underexplored. Here we summarize and evaluate the utility of various diagnostic techniques and the roles of different classes of MET-targeted therapies in cancers with MET amplification, mutation and fusion, and MET overexpression.
Collapse
Affiliation(s)
- Robin Guo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jia Luo
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
290
|
Sholl LM, Hirsch FR, Hwang D, Botling J, Lopez-Rios F, Bubendorf L, Mino-Kenudson M, Roden AC, Beasley MB, Borczuk A, Brambilla E, Chen G, Chou TY, Chung JH, Cooper WA, Dacic S, Lantuejoul S, Jain D, Lin D, Minami Y, Moreira A, Nicholson AG, Noguchi M, Papotti M, Pelosi G, Poleri C, Rekhtman N, Tsao MS, Thunnissen E, Travis W, Yatabe Y, Yoshida A, Daigneault JB, Zehir A, Peters S, Wistuba II, Kerr KM, Longshore JW. The Promises and Challenges of Tumor Mutation Burden as an Immunotherapy Biomarker: A Perspective from the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol 2020; 15:1409-1424. [PMID: 32522712 DOI: 10.1016/j.jtho.2020.05.019] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/11/2020] [Accepted: 05/29/2020] [Indexed: 12/26/2022]
Abstract
Immune checkpoint inhibitor (ICI) therapies have revolutionized the management of patients with NSCLC and have led to unprecedented improvements in response rates and survival in a subset of patients with this fatal disease. However, the available therapies work only for a minority of patients, are associated with substantial societal cost, and may lead to considerable immune-related adverse events. Therefore, patient selection must be optimized through the use of relevant biomarkers. Programmed death-ligand 1 protein expression by immunohistochemistry is widely used today for the selection of programmed cell death protein 1 inhibitor therapy in patients with NSCLC; however, this approach lacks robust sensitivity and specificity for predicting response. Tumor mutation burden (TMB), or the number of somatic mutations derived from next-generation sequencing techniques, has been widely explored as an alternative or complementary biomarker for response to ICIs. In theory, a higher TMB increases the probability of tumor neoantigen production and therefore, the likelihood of immune recognition and tumor cell killing. Although TMB alone is a simplistic surrogate of this complex interplay, it is a quantitative variable that can be relatively readily measured using currently available sequencing techniques. A large number of clinical trials and retrospective analyses, employing both tumor and blood-based sequencing tools, have evaluated the performance of TMB as a predictive biomarker, and in many cases reveal a correlation between high TMB and ICI response rates and progression-free survival. Many challenges remain before the implementation of TMB as a biomarker in clinical practice. These include the following: (1) identification of therapies whose response is best informed by TMB status; (2) robust definition of a predictive TMB cut point; (3) acceptable sequencing panel size and design; and (4) the need for robust technical and informatic rigor to generate precise and accurate TMB measurements across different laboratories. Finally, effective prediction of response to ICI therapy will likely require integration of TMB with a host of other potential biomarkers, including tumor genomic driver alterations, tumor-immune milieu, and other features of the host immune system. This perspective piece will review the current clinical evidence for TMB as a biomarker and address the technical sequencing considerations and ongoing challenges in the use of TMB in routine practice.
Collapse
Affiliation(s)
- Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| | - Fred R Hirsch
- Center for Thoracic Oncology, The Tisch Cancer Institute, New York, New York; Icahn School of Medicine, Mount Sinai Health System, New York, New York
| | - David Hwang
- Department of Laboratory Medicine & Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Johan Botling
- Department of Immunology Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Anja C Roden
- Department of Pathology, Mayo Clinic, Rochester, Minnesota
| | - Mary Beth Beasley
- Icahn School of Medicine, Mount Sinai Health System, New York, New York
| | - Alain Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, New York
| | | | - Gang Chen
- Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | | | - Jin-Haeng Chung
- Seoul National University Bundang Hospital, Seoul, South Korea
| | | | - Sanja Dacic
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Sylvie Lantuejoul
- Université Grenoble Alpes, Grenoble, France; Centre Léon Bérard Unicancer, Lyon, France
| | - Deepali Jain
- All India Institute of Medical Sciences, New Delhi, India
| | - Dongmei Lin
- Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Yuko Minami
- Ibarakihigashi National Hospital, Tokai, Japan
| | - Andre Moreira
- Department of Pathology, Weill Cornell Medicine, New York, New York
| | - Andrew G Nicholson
- Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom; National Heart and Lung Institute, Imperial College, London, United Kingdom
| | | | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| | - Giuseppe Pelosi
- University of Milan, Milan Italy; Department of Oncology and Hemato-Oncology, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Milan Italy
| | - Claudia Poleri
- Oggice of Pathology Consultants, Buenos Aires, Argentina
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, Netherlands
| | - William Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | - Ahmet Zehir
- Oggice of Pathology Consultants, Buenos Aires, Argentina
| | - Solange Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | | | - Keith M Kerr
- Department of Pathology, Aberdffn Royal Infirmary, Aberdffn, United Kingdom
| | - John W Longshore
- Carolinas Pathology Group, Atrium Health, Charlotte, North Carolina
| |
Collapse
|
291
|
Osta BE, Ramalingam SS. RET Fusion: Joining the Ranks of Targetable Molecular Drivers in NSCLC. JTO Clin Res Rep 2020; 1:100050. [PMID: 34589946 PMCID: PMC8474418 DOI: 10.1016/j.jtocrr.2020.100050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 11/17/2022] Open
Abstract
The era of precision medicine has resulted in the identification of a number of genomic alterations that can be targeted with novel therapies. In lung adenocarcinomas, a histology structure that accounts for nearly 50% of all cases of lung cancer, and a number of genomic targets have been linked with effective targeted therapies. For patients with advanced-stage lung adenocarcinomas, molecular testing is now a standard part of diagnostic workup; for patients that have specific driver molecular events, targeted therapies have resulted in substantial improvement in efficacy without excessive toxicity. RET gene fusions are present in approximately 1% to 2% of NSCLC. It is emerging as a new targetable driver for this population. Despite sensitivity to platinum-based chemotherapy and conflicting small reports regarding the efficacy of immune checkpoint inhibitors, there have been limited treatment approaches for this subset of patients. Multiple nonselective RET tyrosine kinase inhibitors exhibited modest anti-RET activity with an increased off-target toxicity profile that often required dose interruption, reduction, or treatment cessation. Recently, novel selective RET inhibitors pralsetinib (BLU-667) and selpercatinib (LOXO-292) have exhibited promising clinical activity with low adverse effect profile in early clinical trials. These new agents are poised to represent a new hope for this special subgroup with unmet needs.
Collapse
Affiliation(s)
- Badi El Osta
- Department of Hematology and Oncology, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia.,Emory University School of Medicine, Atlanta, Georgia
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia.,Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
292
|
Kirchner M, Glade J, Lehmann U, Merkelbach-Bruse S, Hummel M, Lehmann A, Trautmann M, Kumbrink J, Jung A, Dietmaier W, Endris V, Kazdal D, Ploeger C, Evert M, Horst D, Kreipe H, Kirchner T, Wardelmann E, Büttner R, Weichert W, Dietel M, Schirmacher P, Stenzinger A, Pfarr N. NTRK testing: First results of the QuiP-EQA scheme and a comprehensive map of NTRK fusion variants and their diagnostic coverage by targeted RNA-based NGS assays. Genes Chromosomes Cancer 2020; 59:445-453. [PMID: 32319699 DOI: 10.1002/gcc.22853] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/21/2022] Open
Abstract
Gene fusions involving the three neurotrophic tyrosine receptor kinase genes NTRK1, NTRK2, or NTRK3 were identified as oncogenic drivers in many cancer types. Two small molecule inhibitors have been tested in clinical trials recently and require the detection of a NTRK fusion gene prior to therapeutic application. Fluorescence in situ hybridization (FISH) and targeted next-generation sequencing (tNGS) assays are commonly used for diagnostic profiling of gene fusions. In the presented study we applied an external quality assessment (EQA) scheme in order to investigate the suitability of FISH and RNA-/DNA-based tNGS for detection of NTRK fusions in a multinational and multicentric ring trial. In total 27 participants registered for this study. Nine institutions took part in the FISH-based and 18 in the NGS-based round robin test, the latter additionally subdivided into low-input and high-input NGS methods (regarding nucleic acid input). Regardless of the testing method applied, all participants received tumor sections of 10 formalin-fixed and paraffin-embedded (FFPE) tissue blocks for in situ hybridization or RNA/DNA extraction, and the results were submitted via an online questionnaire. For FISH testing, eight of nine (88.8%) participants, and for NGS-based testing 15 of 18 (83.3%) participants accomplished the round robin test successfully. The overall high success rate demonstrates that FISH- and tNGS-based NTRK testing can be well established in a routine diagnostic setting. Complementing this dataset, we provide an updated in silico analysis on the coverage of more than 150 NTRK fusion variants by several commercially available RNA-based tNGS panels.
Collapse
Affiliation(s)
- Martina Kirchner
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Julia Glade
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ulrich Lehmann
- Institute of Pathology, University Hospital Hannover, Hannover, Germany
| | | | - Michael Hummel
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | - Annika Lehmann
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | - Marcel Trautmann
- Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Ludwig Maximilian University (LMU), Munich, Germany
| | - Andreas Jung
- Institute of Pathology, Ludwig Maximilian University (LMU), Munich, Germany
| | - Wolfgang Dietmaier
- Institute of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Kazdal
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carolin Ploeger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Evert
- Institute of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - David Horst
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | - Hans Kreipe
- Institute of Pathology, University Hospital Hannover, Hannover, Germany
| | - Thomas Kirchner
- Institute of Pathology, Ludwig Maximilian University (LMU), Munich, Germany
| | - Eva Wardelmann
- Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich (TUM), Munich, Germany
| | - Manfred Dietel
- Institute of Pathology, Charité University Hospital, Berlin, Germany.,Quality Assurance Initiative in Pathology (QuiP), Berlin, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Nicole Pfarr
- Institute of Pathology, Technical University Munich (TUM), Munich, Germany
| |
Collapse
|
293
|
Schrock AB, Madison R, Rosenzweig M, Allen JM, Erlich RL, Wang SY, Chidiac T, Reddy VS, Riess JW, Yassa AE, Shakir A, Miller VA, Alexander BM, Venstrom J, McGregor K, Ali SM. Patients with NSCLCs Harboring Internal Inversions or Deletion Rearrangements of the ALK Gene Have Durable Responses to ALK Kinase Inhibitors. LUNG CANCER-TARGETS AND THERAPY 2020; 11:33-39. [PMID: 32368168 PMCID: PMC7184117 DOI: 10.2147/lctt.s239675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/24/2020] [Indexed: 12/24/2022]
Abstract
Background ALK fusions are targetable drivers in non-small-cell lung cancer (NSCLC). However, patients with NSCLC harboring ALK rearrangements without a fusion partner identified in DNA have also been shown to respond to ALK inhibitors. We aimed to characterize complex ALK variants that may predict sensitivity to multiple approved ALK inhibitors. Methods Comprehensive genomic profiling (CGP) of DNA isolated from formalin-fixed paraffin-embedded (FFPE) tumor tissue or blood-based circulating tumor DNA was performed for 39,159 NSCLC patients during routine clinical care. For a subset of cases, RNA sequencing was performed, and prior ALK test results and clinical treatment information were collected from treating physicians. Results We queried the Foundation Medicine NSCLC database and identified ALK internal inversions, as well as internal deletions, as the sole ALK rearrangements in 6 (0.02%) and 3 (0.01%) of cases, respectively. In cases with ALK internal inversions, RNA testing identified an EML4-ALK fusion in 2/2 cases evaluated, and 3/3 patients treated with ALK inhibitors had durable responses. A single patient with an ALK internal deletion and clinical data available responded to multiple ALK inhibitors. RNA data available for a subset of non-NSCLC cases suggest that ALK internal deletions removing a portion of the N-terminus are drivers themselves and do not result in ALK fusions. Fluorescence in situ hybridization (FISH) results were inconsistent for both classes of DNA events. Conclusion Rare internal inversions of ALK appear to be indicative of ALK fusions, which can be detected in RNA, and response to ALK inhibitors in patients with NSCLC. In contrast, ALK internal deletions are not associated with ALK fusions in RNA but likely represent targetable drivers themselves. These data suggest that CGP of DNA should be supplemented with immunohistochemistry or RNA-based testing to further resolve these events and match patients to effective therapies.
Collapse
Affiliation(s)
- Alexa B Schrock
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| | - Russell Madison
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| | - Mark Rosenzweig
- Foundation Medicine, Department of Translational Oncology and Clinical Reporting, Cambridge, MA, USA
| | - Justin M Allen
- Foundation Medicine, Department of Translational Oncology and Clinical Reporting, Cambridge, MA, USA
| | - Rachel L Erlich
- Foundation Medicine, Department of Translational Oncology and Clinical Reporting, Cambridge, MA, USA
| | - Siao-Yi Wang
- Loyola University Medical Cancer, Department of Hematology and Oncology, Maywood, IL, USA
| | - Tarek Chidiac
- Zangmeister Cancer Center, Department of Hematology and Oncology, Columbus, OH, USA
| | - Vodur Suresh Reddy
- Cancer Care Specialists, Department of Hematology and Oncology, Reno, NV, USA
| | - Jonathan W Riess
- UC Davis Comprehensive Cancer Center, Department of Hematology and Oncology Sacramento, CA, USA
| | - Ahmet Ersin Yassa
- UC Davis Comprehensive Cancer Center, Department of Hematology and Oncology Sacramento, CA, USA
| | - Abdur Shakir
- Sarah Bush Lincoln Health System, Department of Medical Oncology, Mattoon, IL, USA
| | - Vincent A Miller
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| | - Brian M Alexander
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| | - Jeffrey Venstrom
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| | - Kimberly McGregor
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| | - Siraj M Ali
- Foundation Medicine, Department of Clinical Development, Cambridge, MA, USA
| |
Collapse
|
294
|
Rolfo C, Cardona AF, Cristofanilli M, Paz-Ares L, Diaz Mochon JJ, Duran I, Raez LE, Russo A, Lorente JA, Malapelle U, Gil-Bazo I, Jantus-Lewintre E, Pauwels P, Mok T, Serrano MJ. Challenges and opportunities of cfDNA analysis implementation in clinical practice: Perspective of the International Society of Liquid Biopsy (ISLB). Crit Rev Oncol Hematol 2020; 151:102978. [PMID: 32428812 DOI: 10.1016/j.critrevonc.2020.102978] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Precision medicine was born with the development of new diagnostic techniques and targeted drugs, yielding better outcomes in cancer care. With the evolution and increasing sensitivity for detecting oncogenic drivers, liquid biopsies (LBs), specifically cell-free DNA (cfDNA) analysis, have been proposed as a minimally-invasive technique for genomic profiling. Ranging from sequencing techniques to PCR-based methods and other more complex strategies, this approach, currently applicable in some solid tumors with robust evidence, is showing promising opportunities in other cancers. However, difficulties in validating their clinical utility exist within limitation at different levels among several techniques, reporting of the results, lack of appropriate clinical trial designs, and unknown economic impact. One of the aims of the ISLB is to create recommendations to develop reliable and sustainable diagnostic, prognostic and predictive tools using LBs. This paper is addressing these objectives, helping the healthcare providers and scientific community to understand the potential of LB.
Collapse
Affiliation(s)
- Christian Rolfo
- Thoracic Oncology Department and Early Phase Clinical Trials Section, School of Medicine, Maryland University, Maryland, USA.
| | - Andrés F Cardona
- Clinical and Translational Oncology Group, Clínica del Country, Bogotá, Colombia; Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia; Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, 710 N Fairbanks Court, Suite 8-250A, Chicago, IL, 60611, USA
| | - Luis Paz-Ares
- Hospital Universitario 12 de Octubre, CNIO-H12o Lung Cancer Unit, Universidad Complutense and CIBERONC, Madrid, Spain
| | - Juan Jose Diaz Mochon
- DestiNA Genomica S.L. Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Innovación 1, Edificio BIC, 18016, Armilla, Granada, Spain; GENYO Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government. PTS Granada - Avenida de la Ilustración, 114- 18016, Granada, Spain; Department Medicinal and Organic Chemistry, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain
| | - Ignacio Duran
- Servicio de Oncologia Medica, Medical Oncology Department, Hospital Universitario Marques de Valdecilla, Edificio Sur, 2 Planta, Despacho 277, 39008, Santander, Spain
| | - Luis E Raez
- Memorial Cancer Institute, Memorial Health Care System, Florida International University, Florida, USA
| | - Alessandro Russo
- Thoracic Oncology Department and Early Phase Clinical Trials Section, School of Medicine, Maryland University, Maryland, USA; Medical Oncology Unit A.O. Papardo & Department of Human Pathology, University of Messina, Italy
| | - Jose A Lorente
- Laboratory of Genetic Identification, Department of Legal Medicine, University of Granada, Av. de la Investigación, 11, 18071, Granada, Spain; Centre for Genomics and Oncological Research - GENYO, Pfizer, University of Granada, Andalusian Regional Government, Granada, Spain
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Ignacio Gil-Bazo
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain; University of Navarra, Center for Applied Medical Research, Program of Solid Tumors, Pamplona, Navarra, Spain; Idisna, Navarra Institute for Health Research, Pamplona, Navarra, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Investigación, Valencia General University Hospital, Valencia, Spain
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, & Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Tony Mok
- State Key Laboratory in Oncology in South China, Hong Kong, China
| | - María José Serrano
- Centre for Genomics and Oncological Research - GENYO, Pfizer, University of Granada, Andalusian Regional Government, Granada, Spain; Bio-Health Research Institute (Instituto de Investigación Biosanitaria ibs. GRANADA), Spain; Complejo Hospitalario Universitario Granada (CHUG), Department of Medical Oncology, University of Granada, Granada, Spain.
| | | |
Collapse
|
295
|
Seto K, Masago K, Fujita S, Haneda M, Horio Y, Hida T, Kuroda H, Hosoda W, Okubo KI. Targeted RNA sequencing with touch imprint cytology samples for non-small cell lung cancer patients. Thorac Cancer 2020; 11:1827-1834. [PMID: 32372482 PMCID: PMC7327906 DOI: 10.1111/1759-7714.13460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 01/04/2023] Open
Abstract
Background RNA‐based sequencing is considered ideal for detecting pathogenic fusion‐genes compared to DNA‐based assays and provides valuable information about the relative expression of driver genes. However, RNA from formalin‐fixed paraffin‐embedded tissue has issues with both quantity and quality, making RNA‐based sequencing difficult in clinical practice. Analyzing stamp‐derived RNA with next‐generation sequencing (NGS) can address the above‐mentioned obstacles. In this study, we validated the analytical specifications and clinical performance of our custom panel for RNA‐based assays on the Ion Torrent platform. Methods To evaluate our custom RNA lung panel, we first examined the gene sequences of RNA derived from 35 NSCLC tissues with diverse backgrounds by conventional methods and NGS. Next, we moved to the clinical phase, where clinical samples (all stamp‐derived RNA) were used to examine variants. In the clinical phase we conducted an NGS analysis while simultaneously applying conventional approaches to assess the feasibility and validity of the panel in clinical practice. Results In the prerun phase, all of the variants confirmed with conventional methods were detected by NGS. In the clinical phase, a total of 80 patients were enrolled and 80 tumor specimens were sequenced from February 2018 to December 2018. There were 66 cases in which the RNA concentration was too low to be measured, but sequencing was successful in the vast majority of cases. The concordance between NGS and conventional methods was 95.0%. Conclusions RNA extraction using stamp specimens and panel sequencing by NGS were considered applicable in clinical settings. Key points Significant findings of the study Next‐generation sequencing using RNA from stamp specimens was able to detect driver gene changes in non‐small cell lung cancer including fusion genes with the same accuracy as conventional methods. What this study adds Using RNA from stamp specimens obtained from biopsy increases the number of candidate cases for RNA sequencing in clinical settings.
Collapse
Affiliation(s)
- Katsutoshi Seto
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan.,Department of Thoracic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Shiro Fujita
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Masataka Haneda
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | | | - Toyoaki Hida
- Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| | | | - Waki Hosoda
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Ken-Ichi Okubo
- Department of Thoracic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
296
|
Harada G, Gongora ABL, da Costa CM, Santini FC. TRK Inhibitors in Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2020; 21:39. [DOI: 10.1007/s11864-020-00741-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
297
|
Tsao MS, Yatabe Y. Old Soldiers Never Die: Is There Still a Role for Immunohistochemistry in the Era of Next-Generation Sequencing Panel Testing? J Thorac Oncol 2020; 14:2035-2038. [PMID: 31757371 DOI: 10.1016/j.jtho.2019.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Ming Sound Tsao
- Laboratory Medicine Program, Department of Pathology, University Health Network, Princess Margaret Cancer Centre and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center, Tokyo, Japan
| |
Collapse
|
298
|
Russo A, Lopes AR, McCusker MG, Garrigues SG, Ricciardi GR, Arensmeyer KE, Scilla KA, Mehra R, Rolfo C. New Targets in Lung Cancer (Excluding EGFR, ALK, ROS1). Curr Oncol Rep 2020; 22:48. [PMID: 32296961 DOI: 10.1007/s11912-020-00909-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Over the last two decades, the identification of targetable oncogene drivers has revolutionized the therapeutic landscape of non-small cell lung cancer (NSCLC). The extraordinary progresses made in molecular biology prompted the identification of several rare molecularly defined subgroups. In this review, we will focus on the novel and emerging actionable oncogenic drivers in NSCLC. RECENT FINDINGS Recently, novel oncogene drivers emerged as promising therapeutic targets besides the well-established EGFR mutations, and ALK/ROS1 rearrangements, considerably expanding the list of potential exploitable genetic aberrations. However, the therapeutic algorithm in these patients is far less defined. The identification of uncommon oncogene drivers is reshaping the diagnostic and therapeutic approach to NSCLC. The introduction of novel highly selective inhibitors is expanding the use of targeted therapies to rare and ultra-rare subsets of patients, further increasing the therapeutic armamentarium of advanced NSCLC.
Collapse
Affiliation(s)
- Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.,Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Contrada Papardo, 98158, Messina, Italy
| | - Ana Rita Lopes
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.,Portuguese Institute of Oncology (IPO), Porto, Portugal
| | - Michael G McCusker
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Sandra Gimenez Garrigues
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Giuseppina R Ricciardi
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Contrada Papardo, 98158, Messina, Italy
| | - Katherine E Arensmeyer
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Katherine A Scilla
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.
| |
Collapse
|
299
|
Hensley ML, Chavan SS, Solit DB, Murali R, Soslow R, Chiang S, Jungbluth AA, Bandlamudi C, Srinivasan P, Tap WD, Rosenbaum E, Taylor BS, Donoghue MTA, Hyman DM. Genomic Landscape of Uterine Sarcomas Defined Through Prospective Clinical Sequencing. Clin Cancer Res 2020; 26:3881-3888. [PMID: 32299819 DOI: 10.1158/1078-0432.ccr-19-3959] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/16/2020] [Accepted: 04/10/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE We examined whether prospective molecular characterization of advanced metastatic disease can reveal grade and/or histology-specific differences to inform diagnosis and facilitate enrollment onto clinical trials. EXPERIMENTAL DESIGN Patients with uterine sarcoma consented to a prospective study of next-generation sequencing (NGS). Clinical annotations were extracted from their medical record. Tumor and matched normal DNA were subjected to NGS, and the genomic landscape was explored for survival correlations and therapeutic targetability. RESULTS Tumors from 107 women were sequenced and included leiomyosarcoma (n = 80), high-grade non-leiomyosarcoma (n = 22), low-grade endometrial stromal sarcoma (LG-ESS, n = 4), and smooth muscle tumor of uncertain malignant potential (STUMP, n = 2). Genomic profiling influenced histologic diagnosis in three cases. Common uterine leiomyosarcoma alterations were loss-of-function mutations in TP53 (56%), RB1 (51%), and ATRX (31%). Homozygous deletions of BRCA2 were present in 5% of these patients. PTEN alteration frequency was higher in the metastases samples as compared with the primary samples. Genomes of low-grade tumors were largely silent, while 50.5% of high-grade tumors had whole-genome duplication. Two metastatic uterine leiomyosarcoma cases were hypermutated. Both had prolonged disease-free survival. Potentially actionable mutations were identified in 48 patients (45%), 8 (17%) of whom received matched therapy with 2 achieving clinical responses. Among patients with uterine leiomyosarcoma with somatic BRCA2 alterations, sustained partial responses were observed with PARP inhibitor-containing therapy. DISCUSSION Prospective genomic profiling can contribute to diagnostic precision and inform treatment selection in patients with uterine sarcomas. There was evidence of clinical benefit in patients with uterine leiomyosarcoma with somatic BRCA2 alterations treated with PARP inhibitors.
Collapse
Affiliation(s)
- Martee L Hensley
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| | - Shweta S Chavan
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B Solit
- Weill Cornell Medical College, New York, New York.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rajmohan Murali
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert Soslow
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Chiang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chaitanya Bandlamudi
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Preethi Srinivasan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D Tap
- Weill Cornell Medical College, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evan Rosenbaum
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Weill Cornell Medical College, New York, New York.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark T A Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David M Hyman
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
300
|
Wu L, Deng Q, Xu Z, Zhou S, Li C, Li YX. A novel virtual barcode strategy for accurate panel-wide variant calling in circulating tumor DNA. BMC Bioinformatics 2020; 21:127. [PMID: 32245364 PMCID: PMC7118954 DOI: 10.1186/s12859-020-3412-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/12/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Hybrid capture-based next-generation sequencing of DNA has been widely applied in the detection of circulating tumor DNA (ctDNA). Various methods have been proposed for ctDNA detection, but low-allelic-fraction (AF) variants are still a great challenge. In addition, no panel-wide calling algorithm is available, which hiders the full usage of ctDNA based 'liquid biopsy'. Thus, we developed the VBCALAVD (Virtual Barcode-based Calling Algorithm for Low Allelic Variant Detection) in silico to overcome these limitations. RESULTS Based on the understanding of the nature of ctDNA fragmentation, a novel platform-independent virtual barcode strategy was established to eliminate random sequencing errors by clustering sequencing reads into virtual families. Stereotypical mutant-family-level background artifacts were polished by constructing AF distributions. Three additional robust fine-tuning filters were obtained to eliminate stochastic mutant-family-level noises. The performance of our algorithm was validated using cell-free DNA reference standard samples (cfDNA RSDs) and normal healthy cfDNA samples (cfDNA controls). For the RSDs with AFs of 0.1, 0.2, 0.5, 1 and 5%, the mean F1 scores were 0.43 (0.25~0.56), 0.77, 0.92, 0.926 (0.86~1.0) and 0.89 (0.75~1.0), respectively, which indicates that the proposed approach significantly outperforms the published algorithms. Among controls, no false positives were detected. Meanwhile, characteristics of mutant-family-level noise and quantitative determinants of divergence between mutant-family-level noises from controls and RSDs were clearly depicted. CONCLUSIONS Due to its good performance in the detection of low-AF variants, our algorithm will greatly facilitate the noninvasive panel-wide detection of ctDNA in research and clinical settings. The whole pipeline is available at https://github.com/zhaodalv/VBCALAVD.
Collapse
Affiliation(s)
- Leilei Wu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qinfang Deng
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Ze Xu
- Smartquerier Biomedicine, Shanghai, 201203, China
| | - Songwen Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Chao Li
- Smartquerier Biomedicine, Shanghai, 201203, China.
- Shanghai Center for Bioinformation Technology, Shanghai, 201203, China.
| | - Yi-Xue Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Shanghai Center for Bioinformation Technology, Shanghai, 201203, China.
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|