201
|
Abstract
The detection of gene rearrangements in pediatric leukemia is an essential component of the work-up, with implications for accurate diagnosis, proper risk stratification, and therapeutic decisions, including the use of targeted therapies. The traditional methods of karyotype and fluorescence in situ hybridization are still valuable, but many new assays are also available, with different strengths and weaknesses. These assays include next-generation sequencing-based assays that have the potential for highly multiplexed and/or unbiased detection of rearrangements.
Collapse
Affiliation(s)
- Marian H Harris
- Department of Pathology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
202
|
Wei D, Chen D, Li S, Xiao M, Ren G. A novel DPP10 (intergenic)-NTRK3 fusion in a patient with facial malignant melanoma and potentially benefit from NTRK inhibitors. Oral Oncol 2021; 120:105288. [PMID: 33846089 DOI: 10.1016/j.oraloncology.2021.105288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Dongliang Wei
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 210000, China
| | - Dongsheng Chen
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210002, China
| | - Si Li
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210002, China
| | - Mingzhe Xiao
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210002, China
| | - Guoxin Ren
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 210000, China.
| |
Collapse
|
203
|
Dalurzo ML, Avilés-Salas A, Soares FA, Hou Y, Li Y, Stroganova A, Öz B, Abdillah A, Wan H, Choi YL. Testing for EGFR Mutations and ALK Rearrangements in Advanced Non-Small-Cell Lung Cancer: Considerations for Countries in Emerging Markets. Onco Targets Ther 2021; 14:4671-4692. [PMID: 34511936 PMCID: PMC8420791 DOI: 10.2147/ott.s313669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/08/2021] [Indexed: 12/24/2022] Open
Abstract
The treatment of patients with advanced non-small-cell lung cancer (NSCLC) in recent years has been increasingly guided by biomarker testing. Testing has centered on driver genetic alterations involving the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) rearrangements. The presence of these mutations is predictive of response to targeted therapies such as EGFR tyrosine kinase inhibitors (TKIs) and ALK TKIs. However, there are substantial challenges for the implementation of biomarker testing, particularly in emerging countries. Understanding the barriers to testing in NSCLC will be key to improving molecular testing rates worldwide and patient outcomes as a result. In this article, we review EGFR mutations and ALK rearrangements as predictive biomarkers for NSCLC, discuss a selection of appropriate tests and review the literature with respect to the global uptake of EGFR and ALK testing. To help improve testing rates and unify procedures, we review our experiences with biomarker testing in China, South Korea, Russia, Turkey, Brazil, Argentina and Mexico, and propose a set of recommendations that pathologists from emerging countries can apply to assist with the diagnosis of NSCLC.
Collapse
Affiliation(s)
- Mercedes L Dalurzo
- Department of Pathology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | | | | | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yuan Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Anna Stroganova
- N.N. Blokhin National Medical Research Centre of Oncology, Russian Academy of Medical Sciences, Moscow, Russia
| | - Büge Öz
- Cerrahpaşa School of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Arif Abdillah
- Takeda Pharmaceuticals International AG – Singapore Branch, Singapore, Singapore
| | - Hui Wan
- Takeda Pharmaceuticals International AG – Singapore Branch, Singapore, Singapore
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
204
|
Abstract
The identification of large chromosomal rearrangements in cancers has multiplied exponentially over the last decade. These complex and often rare genomic events have traditionally been challenging to study, in part owing to lack of tools that efficiently engineer disease-associated inversions, deletions and translocations in model systems. The emergence and refinement of genome editing technologies, such as CRISPR, have significantly expanded our ability to generate and interrogate chromosomal aberrations to better understand the networks that govern cancer growth. Here we review how existing technologies are employed to faithfully model cancer-associated chromosome rearrangements in the laboratory, with the ultimate goal of developing more accurate pre-clinical models of and therapeutic strategies for cancers driven by these genomic events.
Collapse
Affiliation(s)
- Salvador Alonso
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Lukas E. Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
205
|
Drilon A, Duruisseaux M, Han JY, Ito M, Falcon C, Yang SR, Murciano-Goroff YR, Chen H, Okada M, Molina MA, Wislez M, Brun P, Dupont C, Branden E, Rossi G, Schrock A, Ali S, Gounant V, Magne F, Blum TG, Schram AM, Monnet I, Shih JY, Sabari J, Pérol M, Zhu VW, Nagasaka M, Doebele R, Camidge DR, Arcila M, Ou SHI, Moro-Sibilot D, Rosell R, Muscarella LA, Liu SV, Cadranel J. Clinicopathologic Features and Response to Therapy of NRG1 Fusion-Driven Lung Cancers: The eNRGy1 Global Multicenter Registry. J Clin Oncol 2021; 39:2791-2802. [PMID: 34077268 PMCID: PMC8407651 DOI: 10.1200/jco.20.03307] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/03/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Although NRG1 fusions are oncogenic drivers across multiple tumor types including lung cancers, these are difficult to study because of their rarity. The global eNRGy1 registry was thus established to characterize NRG1 fusion-positive lung cancers in the largest and most diverse series to date. METHODS From June 2018 to February 2020, a consortium of 22 centers from nine countries in Europe, Asia, and the United States contributed data from patients with pathologically confirmed NRG1 fusion-positive lung cancers. Profiling included DNA-based and/or RNA-based next-generation sequencing and fluorescence in situ hybridization. Anonymized clinical, pathologic, molecular, and response (RECIST v1.1) data were centrally curated and analyzed. RESULTS Although the typified never smoking (57%), mucinous adenocarcinoma (57%), and nonmetastatic (71%) phenotype predominated in 110 patients with NRG1 fusion-positive lung cancer, further diversity, including in smoking history (43%) and histology (43% nonmucinous and 6% nonadenocarcinoma), was elucidated. RNA-based testing identified most fusions (74%). Molecularly, six (of 18) novel 5' partners, 20 unique epidermal growth factor domain-inclusive chimeric events, and heterogeneous 5'/3' breakpoints were found. Platinum-doublet and taxane-based (post-platinum-doublet) chemotherapy achieved low objective response rates (ORRs 13% and 14%, respectively) and modest progression-free survival medians (PFS 5.8 and 4.0 months, respectively). Consistent with a low programmed death ligand-1 expressing (28%) and low tumor mutational burden (median: 0.9 mutations/megabase) immunophenotype, the activity of chemoimmunotherapy and single-agent immunotherapy was poor (ORR 0%/PFS 3.3 months and ORR 20%/PFS 3.6 months, respectively). Afatinib achieved an ORR of 25%, not contingent on fusion type, and a 2.8-month median PFS. CONCLUSION NRG1 fusion-positive lung cancers were molecularly, pathologically, and clinically more heterogeneous than previously recognized. The activity of cytotoxic, immune, and targeted therapies was disappointing. Further research examining NRG1-rearranged tumor biology is needed to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Michael Duruisseaux
- Respiratory Department, Louis Pradel Hospital, Hospices Civils de Lyon Cancer Institute, Lyon, France
- Anticancer Antibodies Laboratory, Cancer Research Center of Lyon, Lyon, France
- Université Claude Bernard Lyon UMR INSERM 1052 CNRS 5286, Université de Lyon, Lyon, France
| | - Ji-Youn Han
- National Cancer Center, Korea, Goyang-si, South Korea
| | - Masaoki Ito
- Pangaea Oncology, Quiron-Dexeus University Institute, Barcelona, Spain
- Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Christina Falcon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | | | - Haiquan Chen
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Morihito Okada
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Miguel Angel Molina
- Pangaea Oncology, Laboratory of Molecular Biology, Quiron-Dexeus University Institute, Barcelona, Spain
| | - Marie Wislez
- Université de Paris, Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Paris, France
- Team Inflammation, Complement, and Cancer, and Oncology Thoracic Unit Pulmonology Department, AP-HP, Hôpital Cochin, Paris, France
| | - Philippe Brun
- Department of Pneumology, Lungenklinik Heckeshorn, Helios Klinikum Emil von Behring, Valence, France
| | - Clarisse Dupont
- Respiratory Department, Louis Pradel Hospital, Hospices Civils de Lyon Cancer Institute, Lyon, France
| | - Eva Branden
- Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden
- Centre for Research and Development, Uppsala University/Region Gävleborg, Gävle, Sweden
| | - Giulio Rossi
- Local Health Authority of Romagna, Infermi Hospital, Rimini, Italy
- Local Health Authority of Romagna, St Maria delle Croci Hospital, Ravenna, Italy
| | | | - Siraj Ali
- Foundation Medicine Inc, Cambridge, MA
| | - Valérie Gounant
- Department of Pulmonology, Hôpital Tenon, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Fanny Magne
- Hopital Nord Ouest Villefranche sur Saône, Gleizé, France
| | | | | | - Isabelle Monnet
- Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jin-Yuan Shih
- National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Joshua Sabari
- New York University Langone Health Perlmutter Cancer Center, New York, NY
| | | | - Viola W. Zhu
- Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology/Oncology, University of California, Irvine School of Medicine, Orange, CA
| | - Misako Nagasaka
- Karmanos Cancer Institute, Wayne State University, Detroit, MI
- Division of Neurology, Department of Internal Medicine, St Marianna University, Kawasaki, Japan
| | - Robert Doebele
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO
| | - D. Ross Camidge
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO
| | - Maria Arcila
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA
| | - Denis Moro-Sibilot
- Clinique de Pneumologie, Pôle Médecine Aiguë Communautaire, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Rafael Rosell
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Stephen V. Liu
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Jacques Cadranel
- Department of Pneumology and Thoracic Oncology, Assistance Publique-Hopitaux de Paris, Tenon Hospital and GRC Theranoscan Sorbonne Université, Paris, France
| |
Collapse
|
206
|
Drenner K, Basu GD, Goodman LJ, Ozols AA, LoBello JR, Royce T, Gordon MS, Borazanci EH, Steinbach MA, Trent J, Sharma S. The value of comprehensive genomic sequencing to maximize the identification of clinically actionable alterations in advanced cancer patients: a case series. Oncotarget 2021; 12:1836-1847. [PMID: 34504655 PMCID: PMC8416559 DOI: 10.18632/oncotarget.28046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023] Open
Abstract
PURPOSE We present seven cases of advanced cancer patients who initially underwent tumor testing utilizing smaller, panel-based tests, followed by a variety of therapeutic treatments which ultimately resulted in progression of their disease. These cases demonstrate the value of utilizing WES/RNA seq and characterization following disease progression in these patients and the determination of clinically targetable alterations as well as acquired resistance mutations. MATERIALS AND METHODS All patients are part of an IRB approved observational study. WES and RNA sequencing were performed, using GEM ExTra® on tumor and blood samples obtained during routine clinical care. To accurately determine somatic versus germline alterations the test was performed with paired normal testing from peripheral blood. RESULTS The presented cases demonstrate the clinical impact of actionable findings uncovered using GEM ExTra® in patients with advanced disease who failed many rounds of treatment. Unique alterations were identified resulting in newly identified potential targeted therapies, mechanisms of resistance, and variation in the genomic characterization of the primary versus the metastatic tumor. CONCLUSIONS Taken together our results demonstrate that GEM ExTra® maximizes detection of actionable mutations, thus allowing for appropriate treatment selection for patients harboring both common and rare genomic alterations.
Collapse
Affiliation(s)
- Kevin Drenner
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
- These authors contributed equally to this work
| | - Gargi D. Basu
- Ashion Analytics, LLC, Phoenix, AZ 85004, USA
- These authors contributed equally to this work
| | | | | | | | | | | | | | | | - Jeffrey Trent
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
| | - Sunil Sharma
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
| |
Collapse
|
207
|
Yao J, Arcila ME, Ladanyi M, Hechtman JF. Pan-Cancer Biomarkers: Changing the Landscape of Molecular Testing. Arch Pathol Lab Med 2021; 145:692-698. [PMID: 33373449 DOI: 10.5858/arpa.2020-0513-ra] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The increasing use of large panel next-generation sequencing technologies in clinical settings has facilitated the identification of pan-cancer biomarkers, which can be diagnostic, prognostic, predictive, or most importantly, actionable. OBJECTIVE.— To discuss recently approved and emerging pan-cancer and multihistology biomarkers as well as testing methodologies. DATA SOURCES.— The US Food and Drug Administration approval documents, National Comprehensive Cancer Network guidelines, literature, and authors' own publications. CONCLUSIONS.— Since 2017, the US Food and Drug Administration has approved genotype-directed therapies for pan-cancer biomarkers, including microsatellite instability, neurotrophic receptor kinases fusions, and high-tumor mutation burden. Both the importance and rarity of these biomarkers have increased the prevalence of genomic profiling across solid malignancies. As an integral part of the management team of patients with advanced cancer, pathologists need to be aware of these emerging biomarkers, the therapies for which they determine eligibility, and the strengths and pitfalls of the available clinical assays.
Collapse
Affiliation(s)
- Jinjuan Yao
- From the Diagnostic Molecular Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- From the Diagnostic Molecular Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- From the Diagnostic Molecular Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaclyn F Hechtman
- From the Diagnostic Molecular Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
208
|
Hasegawa N, Kohsaka S, Kurokawa K, Shinno Y, Takeda Nakamura I, Ueno T, Kojima S, Kawazu M, Suehara Y, Ishijima M, Goto Y, Kojima Y, Yonemori K, Hayashi T, Saito T, Shukuya T, Takahashi F, Takahashi K, Mano H. Highly sensitive fusion detection using plasma cell-free RNA in non-small-cell lung cancers. Cancer Sci 2021; 112:4393-4403. [PMID: 34310819 PMCID: PMC8486187 DOI: 10.1111/cas.15084] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
ALK, ROS1, and RET kinase fusions are important predictive biomarkers of tyrosine kinase inhibitors (TKIs) in non‐small‐cell lung cancer (NSCLC). Analysis of cell‐free DNA (cfDNA) provides a noninvasive method to identify gene changes in tumor cells. The present study sought to use cfRNA and cfDNA for identifying fusion genes. A reliable protocol was established to detect fusion genes using cfRNA and assessed the analytical validity and clinical usefulness in 30 samples from 20 cases of fusion‐positive NSCLC. The results of cfRNA‐based assays were compared with tissue biopsy and cfDNA‐based liquid biopsy (Guardant360 plasma next‐generation sequencing [NGS] assay). The overall sensitivity of the cfRNA‐based assay was 26.7% (8/30) and that of cfDNA‐based assay was 16.7% (3/18). When analysis was limited to the samples collected at chemo‐naïve or progressive disease status and available for both assays, the sensitivity of the cfRNA‐based assay was 77.8% (7/9) and that of cfDNA‐based assay was 33.3% (3/9). Fusion gene identification in cfRNA was correlated with treatment response. These results suggest that the proposed cfRNA assay is a useful diagnostic test for patients with insufficient tissues to facilitate effective administration of first‐line treatment and is a useful tool to monitor the progression of NSCLC for consideration of second‐line treatments.
Collapse
Affiliation(s)
- Nobuhiko Hasegawa
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kana Kurokawa
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ikuko Takeda Nakamura
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Shinya Kojima
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Kojima
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takehito Shukuya
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
209
|
Hou D, Zheng X, Song W, Liu X, Wang S, Zhou L, Tao X, Lv L, Sun Q, Jin Y, Zhang Z, Ding L, Wu N, Zhao S. Association of anaplastic lymphoma kinase variants and alterations with ensartinib response duration in non-small cell lung cancer. Thorac Cancer 2021; 12:2388-2399. [PMID: 34288491 PMCID: PMC8410530 DOI: 10.1111/1759-7714.14083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 01/18/2023] Open
Abstract
Background Here, we aimed to assess the association of ALK variants and alterations with ensartinib response duration in NSCLC, and explore the potential value of computed tomography (CT) radiomic features in predicting progression‐free survival (PFS). Methods We enrolled 88 patients with identified ALK variant NSCLC in a multicenter phase 2 trial, and assessed the impact of ALK variants and secondary ALK alterations on the clinical outcome (response duration) of patients receiving ensartinib. We also established a multifactorial model of clinicopathological and quantitative CT radiomic features to predict PFS and risk stratification. Kaplan–Meier analysis was conducted to identify risk factors for tumor progression. Results Univariate analysis indicated a statistical difference (p = 0.035) in PFS among ALK variants in three classifications (V1, V3, and other variants). Secondary ALK alterations were adversely associated with PFS both in univariate (p = 0.008) and multivariate (p = 0.04) analyses and could identify patients at high risk for early progression in the Kaplan–Meier analysis (p = 0.002). Additionally, response duration to crizotinib <1 year and liver metastasis were adversely associated with PFS. The combined model, composed of clinicopathological signature and CT radiomic signature, showed good prediction ability with the area under the receiver operating characteristic curve being 0.85, and 0.89 in the training and validation dataset respectively. Conclusions Our study showed that secondary ALK alterations were adversely associated with ensartinib efficacy, and that ALK variants might not correlate with PFS. The quantitative radiomic signature provided added prognostic prediction value to the clinicopathological features.
Collapse
Affiliation(s)
- Donghui Hou
- Department of Diagnostic RadiologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaomin Zheng
- Department of EndocrinologyChui Yang Liu Hospital affiliated to Tsinghua UniversityBeijingChina
| | - Wei Song
- Department of RadiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xiaoqing Liu
- Department of Pulmonary OncologyThe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | | | - Lina Zhou
- Department of Diagnostic RadiologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiuli Tao
- PET‐CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lv Lv
- PET‐CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qi Sun
- Department of RadiologyHarbin Medical University Cancer HospitalHarbinChina
| | - Yujing Jin
- PET‐CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zewei Zhang
- PET‐CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | | | - Ning Wu
- Department of Diagnostic RadiologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- PET‐CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shijun Zhao
- Department of Diagnostic RadiologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
210
|
Xia P, Zhang L, Li P, Liu E, Li W, Zhang J, Li H, Su X, Jiang G. Molecular characteristics and clinical outcomes of complex ALK rearrangements identified by next-generation sequencing in non-small cell lung cancers. J Transl Med 2021; 19:308. [PMID: 34271921 PMCID: PMC8283930 DOI: 10.1186/s12967-021-02982-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/11/2021] [Indexed: 11/30/2022] Open
Abstract
Background Complex kinase rearrangement, a mutational process involving one or two chromosomes with clustered rearrangement breakpoints, interferes with the accurate detection of kinase fusions by DNA-based next-generation sequencing (NGS). We investigated the characteristics of complex ALK rearrangements in non-small cell lung cancers using multiple molecular tests. Methods Samples of non-small cell lung cancer patients were analyzed by targeted-capture DNA-based NGS with probes tilling the selected intronic regions of fusion partner genes, RNA-based NGS, RT-PCR, immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). Results In a large cohort of 6576 non-small cell lung cancer patients, 343 (5.2%) cases harboring ALK rearrangements were identified. Fourteen cases with complex ALK rearrangements were identified by DNA-based NGS and classified into three types by integrating various genomic features, including intergenic (n = 3), intragenic (n = 5) and “bridge joint” rearrangements (n = 6). All thirteen cases with sufficient samples actually expressed canonical EML4-ALK fusion transcripts confirmed by RNA-based NGS. Besides, positive ALK IHC was detected in 13 of 13 cases, and 9 of 11 cases were positive in FISH testing. Patients with complex ALK rearrangements who received ALK inhibitors treatment (n = 6), showed no difference in progression-free survival (PFS) compared with patients with canonical ALK fusions n = 36, P = 0.9291). Conclusions This study firstly reveals the molecular characteristics and clinical outcomes of complex ALK rearrangements in NSCLC, sensitive to ALK inhibitors treatment, and highlights the importance of utilizing probes tilling the selected intronic regions of fusion partner genes in DNA-based NGS for accurate fusion detection. RNA and protein level assay may be critical in validating the function of complex ALK rearrangements in clinical practice for optimal treatment decision. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02982-4.
Collapse
Affiliation(s)
- Peiyi Xia
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Jian She Dong Road 1, Zhengzhou, 450052, Henan, China
| | - Lan Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Jian She Dong Road 1, Zhengzhou, 450052, Henan, China
| | - Pan Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Jian She Dong Road 1, Zhengzhou, 450052, Henan, China
| | - Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Jian She Dong Road 1, Zhengzhou, 450052, Henan, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Jian She Dong Road 1, Zhengzhou, 450052, Henan, China
| | - Jianying Zhang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Hui Li
- Clinical Research Division, Berry Oncology Corporation, Fuzhou, 350200, China
| | - Xiaoxing Su
- Clinical Research Division, Berry Oncology Corporation, Fuzhou, 350200, China
| | - Guozhong Jiang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Jian She Dong Road 1, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
211
|
Zhang W, Yao J, Zhong M, Zhang Y, Guo X, Wang HY. A Brief Overview and Update on Major Molecular Genomic Alterations in Solid, Bone and Soft Tissue Tumors, Hematopoietic As Well As Lymphoid Malignancies. Arch Pathol Lab Med 2021; 145:1358-1366. [PMID: 34270703 DOI: 10.5858/arpa.2021-0077-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Recent advances in comprehensive genomic profiling by next-generation sequencing have uncovered the genomic alterations at the molecular level for many types of tumors; as such, numerous small specific molecules that target these alterations have been developed and widely used in the management of these cancers. OBJECTIVE.— To provide a concise molecular genomic update in solid, bone and soft tissue tumors, hematopoietic as well as lymphoid malignancies; discuss its clinical applications; and familiarize practicing pathologists with the emerging cancer biomarkers and their diagnostic utilities. DATA SOURCES.— This review is based on the National Comprehensive Cancer Network guidelines and peer-reviewed English literature. CONCLUSIONS.— Tumor-specific biomarkers and molecular/genomic alterations, including pan-cancer markers, have been significantly expanded in the past decade thanks to large-scale high-throughput technologies and will continue to emerge in the future. These biomarkers can be of great value in diagnosis, prognosis, and/or targeted therapy/treatment. Familiarization with these emerging and ever-changing tumor biomarkers will undoubtedly aid pathologists in making accurate and state-of-the-art diagnoses and enable them to be more actively involved in the care of cancer patients.
Collapse
Affiliation(s)
- Wei Zhang
- From the Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison (W. Zhang).,W. Zhang and Yao are co-first authors.,W. Zhang and H.-Y. Wang are co-senior authors and supervised this manuscript equally
| | - Jinjuan Yao
- The Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (Yao).,W. Zhang and Yao are co-first authors
| | - Minghao Zhong
- The Department of Pathology, Yale University School of Medicine, New Haven, Connecticut (Zhong)
| | - Yaxia Zhang
- The Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, New York (Y. Zhang).,The Department of Pathology and Laboratory Medicine, Weill Cornell College of Medicine, New York, New York (Y. Zhang)
| | - Xiaoling Guo
- The Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (Guo)
| | - Huan-You Wang
- The Department of Pathology, University of California San Diego, La Jolla (Wang).,W. Zhang and H.-Y. Wang are co-senior authors and supervised this manuscript equally
| |
Collapse
|
212
|
Chang JC, Offin M, Falcon C, Brown D, Houck-Loomis BR, Meng F, Rudneva VA, Won HH, Amir S, Montecalvo J, Desmeules P, Kadota K, Adusumilli PS, Rusch VW, Teed S, Sabari JK, Benayed R, Nafa K, Borsu L, Li BT, Schram AM, Arcila ME, Travis WD, Ladanyi M, Drilon A, Rekhtman N. Comprehensive Molecular and Clinicopathologic Analysis of 200 Pulmonary Invasive Mucinous Adenocarcinomas Identifies Distinct Characteristics of Molecular Subtypes. Clin Cancer Res 2021; 27:4066-4076. [PMID: 33947695 PMCID: PMC8282731 DOI: 10.1158/1078-0432.ccr-21-0423] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/27/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE Invasive mucinous adenocarcinoma (IMA) is a unique subtype of lung adenocarcinoma, characterized genomically by frequent KRAS mutations or specific gene fusions, most commonly involving NRG1. Comprehensive analysis of a large series of IMAs using broad DNA- and RNA-sequencing methods is still lacking, and it remains unclear whether molecular subtypes of IMA differ clinicopathologically. EXPERIMENTAL DESIGN A total of 200 IMAs were analyzed by 410-gene DNA next-generation sequencing (MSK-IMPACT; n = 136) or hotspot 8-oncogene genotyping (n = 64). Driver-negative cases were further analyzed by 62-gene RNA sequencing (MSK-Fusion) and those lacking fusions were further tested by whole-exome sequencing and whole-transcriptome sequencing (WTS). RESULTS Combined MSK-IMPACT and MSK-Fusion testing identified mutually exclusive driver alterations in 96% of IMAs, including KRAS mutations (76%), NRG1 fusions (7%), ERBB2 alterations (6%), and other less common events. In addition, WTS identified a novel NRG2 fusion (F11R-NRG2). Overall, targetable gene fusions were identified in 51% of KRAS wild-type IMAs, leading to durable responses to targeted therapy in some patients. Compared with KRAS-mutant IMAs, NRG1-rearranged tumors exhibited several more aggressive characteristics, including worse recurrence-free survival (P < 0.0001). CONCLUSIONS This is the largest molecular study of IMAs to date, where we demonstrate the presence of a major oncogenic driver in nearly all cases. This study is the first to document more aggressive characteristics of NRG1-rearranged IMAs, ERBB2 as the third most common alteration, and a novel NRG2 fusion in these tumors. Comprehensive molecular testing of KRAS wild-type IMAs that includes fusion testing is essential, given the high prevalence of alterations with established and investigational targeted therapies in this subset.
Collapse
Affiliation(s)
- Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Offin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina Falcon
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David Brown
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Houck-Loomis
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fanli Meng
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vasilisa A Rudneva
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Helen H Won
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sharon Amir
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph Montecalvo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Patrice Desmeules
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kyuichi Kadota
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Surgery, Thoracic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasad S Adusumilli
- Department of Surgery, Thoracic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valerie W Rusch
- Department of Surgery, Thoracic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Teed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell Biology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua K Sabari
- 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
| | - Khedoudja Nafa
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bob T Li
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alison M Schram
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D Travis
- 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
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Early Drug Development Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
213
|
Zhao R, Yao F, Xiang C, Zhao J, Shang Z, Guo L, Ding W, Ma S, Yu A, Shao J, Zhu L, Han Y. Identification of NTRK gene fusions in lung adenocarcinomas in the Chinese population. J Pathol Clin Res 2021; 7:375-384. [PMID: 33768710 PMCID: PMC8185368 DOI: 10.1002/cjp2.208] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/30/2022]
Abstract
The molecular profile of neurotrophic tyrosine kinase receptor (NTRK) gene fusions in lung adenocarcinoma (LUAD) is not fully understood. Next-generation sequencing (NGS) and pan-tyrosine kinase receptor (TRK) immunohistochemistry (IHC) are powerful tools for NTRK fusion detection. In this study, a total of 4,619 LUAD formalin-fixed, paraffin-embedded tissues were collected from patients who underwent biopsy or resection at the Shanghai Chest Hospital during 2017-2019. All specimens were screened for NTRK1 rearrangements using DNA-based NGS. Thereafter, the cases with NTRK1 rearrangements and cases negative for common driver mutations were analyzed for NTRK1/2/3 fusions using total nucleic acid (TNA)-based NGS and pan-TRK IHC. Overall, four NTRK1/2 fusion events were identified, representing 0.087% of the original sample set. At the DNA level, seven NTRK1 rearrangements were identified, while only two TPM3-NTRK1 fusions were confirmed on TNA-based NGS as functional. In addition, two NTRK2 fusions (SQSTM1-NTRK2 and KIF5B-NTRK2) were identified by TNA-based NGS in 350 'pan-negative' cases. Two patients harboring NTRK1/2 fusions were diagnosed with invasive adenocarcinoma, while the other two were diagnosed with adenocarcinoma in situ and minimally invasive adenocarcinoma. All four samples with NTRK fusions were positive for the expression of pan-TRK. The two samples with NTRK2 fusions showed cytoplasmic staining alone, while the other two samples with NTRK1 fusions exhibited both cytoplasmic and membranous staining. In summary, functional NTRK fusions are found in early-stage LUAD; however, they are extremely rare. According to this study's results, they are independent oncogenic drivers, mutually exclusive with other driver mutations. We demonstrated that NTRK rearrangement analysis using a DNA-based approach should be verified with an RNA-based assay.
Collapse
Affiliation(s)
- Ruiying Zhao
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Chan Xiang
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Jikai Zhao
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Zhanxian Shang
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Lianying Guo
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Wenjie Ding
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Shengji Ma
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Anbo Yu
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Jinchen Shao
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Lei Zhu
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiPR China
| |
Collapse
|
214
|
Slotkin EK, Bowman AS, Levine MF, Dela Cruz F, Coutinho DF, Sanchez GI, Rosales N, Modak S, Tap WD, Gounder MM, Thornton KA, Bouvier N, You D, Gundem G, Gerstle JT, Heaton TE, LaQuaglia MP, Wexler LH, Meyers PA, Kung AL, Papaemmanuil E, Zehir A, Ladanyi M, Shukla N. Comprehensive Molecular Profiling of Desmoplastic Small Round Cell Tumor. Mol Cancer Res 2021; 19:1146-1155. [PMID: 33753552 PMCID: PMC8293793 DOI: 10.1158/1541-7786.mcr-20-0722] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022]
Abstract
Desmoplastic small round cell tumor (DSRCT) is characterized by the EWSR1-WT1 t(11;22) (p13:q12) translocation. Few additional putative drivers have been identified, and research has suffered from a lack of model systems. Next-generation sequencing (NGS) data from 68 matched tumor-normal samples, whole-genome sequencing data from 10 samples, transcriptomic and affymetrix array data, and a bank of DSRCT patient-derived xenograft (PDX) are presented. EWSR1-WT1 fusions were noted to be simple, balanced events. Recurrent mutations were uncommon, but were noted in TERT (3%), ARID1A (6%), HRAS (5%), and TP53 (3%), and recurrent loss of heterozygosity (LOH) at 11p, 11q, and 16q was identified in 18%, 22%, and 34% of samples, respectively. Comparison of tumor-normal matched versus unmatched analysis suggests overcalling of somatic mutations in prior publications of DSRCT NGS data. Alterations in fibroblast growth factor receptor 4 (FGFR4) were identified in 5 of 68 (7%) of tumor samples, whereas differential overexpression of FGFR4 was confirmed orthogonally using 2 platforms. PDX models harbored the pathognomic EWSR1-WT1 fusion and were highly representative of corresponding tumors. Our analyses confirm DSRCT as a genomically quiet cancer defined by the balanced translocation, t(11;22)(p13:q12), characterized by a paucity of secondary mutations but a significant number of copy number alterations. Against this genomically quiet background, recurrent activating alterations of FGFR4 stood out, and suggest that this receptor tyrosine kinase, also noted to be highly expressed in DSRCT, should be further investigated. Future studies of DSRCT biology and preclinical therapeutic strategies should benefit from the PDX models characterized in this study. IMPLICATIONS: These data describe the general quiescence of the desmoplastic small round cell tumor (DSRCT) genome, present the first available bank of DSRCT model systems, and nominate FGFR4 as a key receptor tyrosine kinase in DSRCT, based on high expression, recurrent amplification, and recurrent activating mutations.
Collapse
Affiliation(s)
- Emily K Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Anita S Bowman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Max F Levine
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Filemon Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Diego F Coutinho
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Glorymar I Sanchez
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nestor Rosales
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mrinal M Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine A Thornton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nancy Bouvier
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunes Gundem
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin T Gerstle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Todd E Heaton
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael P LaQuaglia
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leonard H Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul A Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- 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
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
215
|
Yang SR, Chang JC, Leduc C, Tan KS, Dogan S, Benayed R, Borsu L, Offin M, Drilon A, Travis WD, Arcila ME, Ladanyi M, Rekhtman N. Invasive Mucinous Adenocarcinomas With Spatially Separate Lung Lesions: Analysis of Clonal Relationship by Comparative Molecular Profiling. J Thorac Oncol 2021; 16:1188-1199. [PMID: 33839364 PMCID: PMC8240964 DOI: 10.1016/j.jtho.2021.03.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Pulmonary invasive mucinous adenocarcinomas (IMAs) often present with spatially separate lung lesions. Clonal relationship between such lesions, particularly those involving contralateral lobes, is not well established. Here, we used comparative genomic profiling to address this question. METHODS Patients with genomic analysis performed on two IMAs located in different lung regions were identified. Molecular assays included DNA-based next-generation sequencing (NGS) for 410 to 468 genes (Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets), RNA-based NGS for 62 genes (Memorial Sloan Kettering-Fusion), or non-NGS assays. RESULTS Comparative genomic profiling was performed on two separate IMAs in 24 patients, of whom 19 had contralateral lesions. Tumors from all but one patient shared matching driver alterations, including KRAS (n = 19), NRG1 (n = 2), ERBB2 (n = 1) or BRAF (n = 1). In addition, in patients with paired tumors profiled by NGS (n = 12), shared driver alterations were accompanied by up to 4 (average 2.6) other identical mutations, further supporting the clonal relationship between the tumors. Only in a single patient separate IMAs harbored entirely nonoverlapping mutation profiles, supporting clonally unrelated, distinct primary tumors. Notably, in a subset of patients (n = 3), molecular testing confirmed a clonal relationship between the original resected IMAs and subsequent contralateral IMA presenting after an extremely long latency (8.1-11.7 y). CONCLUSIONS Comparative molecular profiling supports that nearly all separate pulmonary IMA lesions represent intrapulmonary spread arising from a single tumor and documents a subset with a remarkably protracted course of intrapulmonary progression. This study reinforces the unique biology and clinical behavior of IMAs while further highlighting the value of genomic testing for clarifying the clonal relationship between multiple lung carcinomas.
Collapse
Affiliation(s)
- Soo-Ryum Yang
- 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
| | - Charles Leduc
- Department of Pathology and Cellular Biology, University of Montreal, Montreal, Quebec, Canada
| | - Kay See Tan
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Snjezana Dogan
- 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
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- 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
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
216
|
Abstract
The genetic basis for pediatric acute myeloid leukemia (AML) is highly heterogeneous, often involving the cooperative action of characteristic chromosomal rearrangements and somatic mutations in progrowth and antidifferentiation pathways that drive oncogenesis. Although some driver mutations are shared with adult AML, many genetic lesions are unique to pediatric patients, and their appropriate identification is essential for patient care. The increased understanding of these malignancies through broad genomic studies has begun to risk-stratify patients based on their combinations of genomic alterations, a trend that will enable precision medicine in this population.
Collapse
Affiliation(s)
- Bryan Krock
- Caris Life Sciences, 4610 South 44th Place, Phoenix, AZ, USA
| | | |
Collapse
|
217
|
Kuang Y, Xu P, Wang J, Zheng Y, Sun X, Li Z, Gan R, Li H, Guo Y, Yao F, Zhu C, Ke Z, Tang K. Detecting ALK Rearrangement with RT-PCR: A Reliable Approach Compared with Next-Generation Sequencing in Patients with NSCLC. Mol Diagn Ther 2021; 25:487-494. [PMID: 34133003 PMCID: PMC8249291 DOI: 10.1007/s40291-021-00532-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 11/26/2022]
Abstract
Background Precise detection of anaplastic lymphoma kinase (ALK) rearrangement guides the application of ALK-targeted tyrosine kinase inhibitors (ALK-TKIs) in patients with non-small-cell lung cancer (NSCLC). Next-generation sequencing (NGS) has been widely used in clinics, but DNA-based NGS used to detect fusion genes has delivered false-negative results. However, fusion genes can be successfully detected at the transcription level and with higher sensitivity using RNA-based reverse transcription polymerase chain reaction (RT-PCR). Objective This study compared the performance of RT-PCR and NGS in the detection of echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusion in Chinese patients with NSCLC. Methods Formalin-fixed paraffin-embedded tissues from 153 patients who were pathologically diagnosed as having NSCLC were collected from November 2017 to October 2019. Both DNA/RNA-based NGS and RNA-based RT-PCR were used to detect EML4-ALK fusion. For samples with discordant ALK status results, fluorescence in situ hybridization (FISH) or Sanger sequencing was used to further confirm the ALK status. Results In total, 124 samples were successfully analyzed using both NGS and RT-PCR. For 118 samples, results were consistent between NGS and RT-PCR, with 25 reported as ALK fusion positive and 93 as ALK fusion negative, achieving a concordance rate of 95.16%. Among the six samples with disconcordant results, five were positive using RT-PCR but negative using NGS, and one was positive using NGS but negative using RT-PCR. Four of six cases with disconcordant results (three RT-PCR positive and one NGS positive) were successfully validated using either FISH or Sanger sequencing. Conclusions Compared with NGS, RT-PCR appears to be a reliable method of detecting EML4-ALK fusion in patients with NSCLC. Supplementary Information The online version contains supplementary material available at 10.1007/s40291-021-00532-8.
Collapse
Affiliation(s)
- Yukun Kuang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Peihang Xu
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Jiyu Wang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yifan Zheng
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue Sun
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Zimu Li
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - RunJing Gan
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Huixia Li
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yubiao Guo
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China
| | - Fei Yao
- Amoy Diagnostics Co., Ltd., Xiamen, China
| | | | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Kejing Tang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China. .,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
218
|
Lee M, Jain P, Wang F, Ma PC, Borczuk A, Halmos B. MET alterations and their impact on the future of non-small cell lung cancer (NSCLC) targeted therapies. Expert Opin Ther Targets 2021; 25:249-268. [PMID: 33945380 DOI: 10.1080/14728222.2021.1925648] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: The MET gene and its pathway normally plays a crucial role in cell homeostasis, motility, and apoptosis. However, when the MET gene is altered, there is an imbalance toward cell proliferation and invasion commonly seen in numerous different types of cancers. The heterogeneous group of MET alterations that includes MET amplification, MET exon 14 skipping mutation, and MET fusions has been difficult to diagnose and treat. Currently, treatments are focused on tyrosine kinase inhibitors but now there is emerging data on novel MET-targeted therapies including monoclonal antibodies and antibody-drug conjugates that have emerged.Areas covered: We introduce new emerging data on MET alterations in non-small cell lung cancer (NSCLC) that has contributed to advances in MET targeted therapeutics. We offer our perspective and examine new information on the mechanisms of the MET alterations in this review.Expert opinion: Given the trends currently involving the targeting of MET altered malignancies, there will most likely be a continued rapid expansion of testing, novel tyrosine kinase inhibitors and potent antibody approaches. Combination treatments will be necessary to optimize management of advanced and early disease.
Collapse
Affiliation(s)
- Matthew Lee
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Prantesh Jain
- Division of Medical Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Feng Wang
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Patrick C Ma
- Penn State CancerInstitute, PennState College of Medicine, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Alain Borczuk
- Department of Pathology, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
219
|
Zhang J, Huang J, Li Q, Lin H, Luo Z, Chen R. DNA and RNA sequencing revealed a complex intergenic-ALK fusion in a lung adenocarcinoma patient who responded to TKI therapy. Lung Cancer 2021; 159:171-174. [PMID: 34266695 DOI: 10.1016/j.lungcan.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Jiexia Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| | - Jian Huang
- Department of Thoracic Surgery, Maoming People's Hospital, Maoming, Guangdong, PR China
| | - Qin Li
- Geneplus-Beijing, Beijing, PR China
| | - Huaming Lin
- The First Tumor Department, Maoming People's Hospital, Maoming, Guangdong, PR China
| | - Zhiqiang Luo
- Department of Thoracic Surgery, Maoming People's Hospital, Maoming, Guangdong, PR China
| | | |
Collapse
|
220
|
What Is New in Biomarker Testing at Diagnosis of Advanced Non-Squamous Non-Small Cell Lung Carcinoma? Implications for Cytology and Liquid Biopsy. JOURNAL OF MOLECULAR PATHOLOGY 2021. [DOI: 10.3390/jmp2020015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The discovery and clinical validation of biomarkers predictive of the response of non-squamous non-small-cell lung carcinomas (NS-NSCLC) to therapeutic strategies continue to provide new data. The evaluation of novel treatments is based on molecular analyses aimed at determining their efficacy. These tests are increasing in number, but the tissue specimens are smaller and smaller and/or can have few tumor cells. Indeed, in addition to tissue samples, complementary cytological and/or blood samples can also give access to these biomarkers. To date, it is recommended and necessary to look for the status of five genomic molecular biomarkers (EGFR, ALK, ROS1, BRAFV600, NTRK) and of a protein biomarker (PD-L1). However, the short- and more or less long-term emergence of new targeted treatments of genomic alterations on RET and MET, but also on others’ genomic alteration, notably on KRAS, HER2, NRG1, SMARCA4, and NUT, have made cellular and blood samples essential for molecular testing. The aim of this review is to present the interest in using cytological and/or liquid biopsies as complementary biological material, or as an alternative to tissue specimens, for detection at diagnosis of new predictive biomarkers of NS-NSCLC.
Collapse
|
221
|
Rodríguez‐Antolín C, Rosas‐Alonso R, Cruz P, Higuera O, Sánchez‐Cabrero D, Esteban‐Rodríguez I, Peláez‐García A, Fernández Montaño VE, Rodríguez‐Jiménez C, Ibáñez de Cáceres I, de Castro J. Novel SLC12A2-ROS1 Fusion in Non-Small Cell Lung Cancer with a Significant Response to Crizotinib: The Importance of Choosing the Appropriate Next-Generation Sequencing Assay. Oncologist 2021; 26:e908-e912. [PMID: 33682977 PMCID: PMC8176992 DOI: 10.1002/onco.13745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/25/2021] [Indexed: 01/05/2023] Open
Abstract
Identifying the druggable target is crucial for patients with nonsquamous advanced non-small cell lung cancer (NSCLC). This article adds to the spectrum of ROS1 fusion cases described in NSCLC. We describe a novel SLC12A2-ROS1 rearrangement that has not been previously reported in other cancers: a fusion that has clinical and radiological sensitivity to crizotinib. Fluorescence in situ hybridization detected the SLC12A2-ROS1 fusion and it was confirmed through hybrid capture-based next-generation sequencing (NGS); however, the fusion could not be detected by amplicon-based assay. The success of implementing NGS into routine clinical practice depends on the accuracy of testing. The test's methodological features should then be considered because they significantly affect the results. Given this patient's response to crizotinib, identifying patients with undescribed ROS1 fusions has important therapeutic implications. KEY POINTS: This is the first known description of an SLC12A2-ROS1 fusion. Considering the patient's clinical features and tumor response observed after crizotinib therapy, the authors confirm that this new rearrangement has relevant clinical impact for patients with non-small cell lung cancer. The success of implementing next-generation sequencing (NGS) into routine clinical practice depends on the accuracy of the testing. Different assays and NGS platforms can achieve differing results. Each assay's limitations need to be considered to ensure the quality of precision medicine in clinical practice.
Collapse
Affiliation(s)
- Carlos Rodríguez‐Antolín
- Epigenetics Laboratory, Department of Genetics, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | - Rocío Rosas‐Alonso
- Epigenetics Laboratory, Department of Genetics, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | - Patricia Cruz
- Department of Medical Oncology, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | - Oliver Higuera
- Department of Medical Oncology, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | - Darío Sánchez‐Cabrero
- Department of Medical Oncology, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | - Isabel Esteban‐Rodríguez
- Department of Medical Oncology, Hospital Universitario La PazMadridSpain
- Department of Pathology, Hospital Universitario La PazMadridSpain
| | - Alberto Peláez‐García
- Molecular Pathology and Therapeutic Targets, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | | | - Carmen Rodríguez‐Jiménez
- Genetics of Metabolic Diseases, Department of Genetics, Hospital Universitario La PazMadridSpain
| | - Inmaculada Ibáñez de Cáceres
- Epigenetics Laboratory, Department of Genetics, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | - Javier de Castro
- Department of Medical Oncology, Hospital Universitario La PazMadridSpain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital Universitario La Paz Institute for Health Research (IdiPAZ)MadridSpain
| |
Collapse
|
222
|
Odintsov I, Somwar R, Ladanyi M, Drilon A. ROS1 at the Crossroads of Clinical Oncology, Molecular Diagnostics, and Drug Development. JCO Oncol Pract 2021; 17:15-16. [PMID: 33434449 DOI: 10.1200/op.20.00969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Romel Somwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, 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
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
223
|
Subramanian J, Tawfik O. Detection of MET exon 14 skipping mutations in non-small cell lung cancer: overview and community perspective. Expert Rev Anticancer Ther 2021; 21:877-886. [PMID: 33957836 DOI: 10.1080/14737140.2021.1924683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Non-small cell lung cancer (NSCLC), which accounts for the majority of lung cancer diagnoses in the United States, has many known driver mutations, including MET exon 14 skipping mutation (METex14). The detection of oncogenic driver mutations in NSCLC and the development of drugs to target these alterations, including METex14, has created the need for accurate and reliable testing, of which next-generation sequencing (NGS) is the gold standard. However, detection of METex14 in patients with NSCLC can be challenging due to the complex biology of METex14 and the abilities of different NGS platforms to detect METex14.Areas covered: This review provides an overview of METex14 biology, discusses the optimal platforms for the detection of METex14 in NSCLC, and provides an overview of the use of NGS in the community setting.Expert opinion: Broad molecular testing is crucial for identifying actionable oncogenic drivers in NSCLC. METex14 is a complex oncogenic driver mutation requiring carefully optimized platforms for proper detection. To identify patients eligible for targeted therapies - including therapies targeting novel oncogenic drivers, such as MET inhibitors - community oncologists need to be aware of both the use of NGS platforms and the differences in their capabilities to detect certain oncogenic drivers.
Collapse
Affiliation(s)
- Janakiraman Subramanian
- Department of Medicine, University of Missouri-Kansas City, School of Medicine, Kansas City, Missouri, USA.,Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri, USA
| | - Ossama Tawfik
- Department of Pathology, Saint Luke's Health System of Kansas City, MAWD Pathology Group, Lenexa, Kansas, USA
| |
Collapse
|
224
|
Wang B, Chen R, Wang C, Guo J, Yuan M, Chen H, Xia X, Zhong D. Identification of novel ALK fusions using DNA/RNA sequencing in immunohistochemistry / RT-PCR discordant NSCLC patients. Hum Pathol 2021; 114:90-98. [PMID: 34019866 DOI: 10.1016/j.humpath.2021.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022]
Abstract
Anaplastic lymphocyte kinase (ALK) rearrangement, a key oncogenic driver promoting the expression of ALK protein in tumor cells, is found in 2%-7% of patients with nonsmall cell lung cancer (NSCLC). ALK fusion is routinely determined with immunohistochemistry (IHC) or RT-PCR in many laboratories. However, there were discordant cases. In this study, we employed a hybridization-based next-generation sequencing (NGS) of DNA and RNA to explore the underlying mechanisms. FFPE tissues of 302 NSCLC tumors, which had been ALK tested with IHC and RT-PCR, were retrospectively studied, of which 18 were IHC positive, and 14 were RT-PCR positive. This resulted in 4 discordant cases, which were further analyzed with NGS. One sample failed the RNA quality control due to extensive RNA degradation. Three non-EML4-ALK fusions were identified in the 4 cases with DNA sequencing, including a CLTC-ALK fusion (EX31:EX19), a WDPCP-ALK fusion (EX14:EX20), and a novel PLB1-ALK fusion (EX6:EX20). Interestingly, two additional fusions: STRN-ALK fusion (EX3:EX20) and DCTN1-ALK fusion (EX20:EX20), were identified with RNA sequencing. The discordance of IHC/RT-PCR was mainly due to limited coverage of non-EML4-ALK fusions in the RT-PCR assay. NGS-based DNA/RNA sequencing appears to be a promising rescue technique for nonclear-cut IHC/RT-PCR cases and also offers a unique opportunity to identify novel ALK fusions.
Collapse
Affiliation(s)
- Bei Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | | | | | - Jia Guo
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | | | - Huang Chen
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | | | - Dingrong Zhong
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
225
|
He Y, Chen L, Zhao L, Dang S, Liu G, Sasada S, Ma PC, van Zandwijk N, Rosell R, Popper HH, Wang H, Jiang M, Guo H, Liu X, Chen S, Zhang X, Xu M, Zhu B, Liu M, Zhou C. Genomic and transcriptional alterations in first-line chemotherapy exert a potentially unfavorable influence on subsequent immunotherapy in NSCLC. Am J Cancer Res 2021; 11:7092-7109. [PMID: 34093873 PMCID: PMC8171101 DOI: 10.7150/thno.58039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/21/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Recent studies in non-small cell lung cancer (NSCLC) patients have demonstrated that first-line immunotherapy is associated with better therapeutic response than second-line treatment. So far, the mechanisms need to be explored. It prompted us to evaluate the association between first-line chemotherapy and subsequent immunotherapy in NSCLC as well as its underlying mechanisms at the genomic and transcriptomic level. Methods: We launched a prospective, observational clinical study, paired tumor biopsies before and after chemotherapy were collected from NSCLC patients without tyrosine kinase inhibitor (TKI)-related driver gene mutations. The analyses included genomic and transcriptional changes performed by next-generation sequencing (NGS)-based whole-exome sequencing (WES) and messager ribonucleic acid (mRNA) sequencing. Characteristic mutational alterations in 1574 genes were investigated based on mutational status, clinicopathological factors, and chemotherapy responses. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, neoantigen prediction and intratumoral heterogeneity evaluation were also performed. Results: Samples and information from 32 NSCLC patients without TKI-related driver gene mutations were obtained. We found that the total number of single nucleotide variants (SNV)/insertion-deletion (INDEL) mutations did not change significantly after chemotherapy. The tumor mutation burden (TMB) decreased significantly after chemotherapy in smoking patients and the decreased TMB correlated with a better survival of smoking patients. The change in copy number variations (CNVs) exhibited a decreasing trend during chemotherapy. Subsequent analysis at mRNA level revealed a significant decrease in the expression levels of genes related to antigen processing and presentation as well as other factors relevant for response to immunotherapy. Pathway enrichment analysis confirmed that the immune-related signaling pathways or biological processes were decreased after first-line chemotherapy. Conclusions: Our study presents an explanation for the unsatisfactory results of immunotherapy when given after chemotherapy, and suggests that first-line chemotherapy is able to influence the tumor microenvironment and decrease the efficacy of subsequent immunotherapy. The study was registered at ClinicalTrials.gov, number NCT03764917, and has completed enrolment; patients are still in follow-up.
Collapse
|
226
|
Abstract
NRG1 fusions are rare oncogenic drivers that exist at low frequencies across multiple tumor types. They are uncommon in lung cancer with an estimated incidence of 0.2 %. NRG1 fusions have a unique biology and are challenging to detect, due to large intronic regions of the gene, but they do represent possible therapeutic targets. Several agents targeting the ErbB signaling pathway have shown early evidence of efficacy including pan-ErbB kinase inhibitors, monoclonal antibodies, and bispecific antibodies. Supporting data are limited to case reports and small series for now, but prospective trials are underway. While our understanding of these fusions is still evolving, it is clear that NRG1 will be a clinically relevant finding in the years to come.
Collapse
Affiliation(s)
- Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.
| |
Collapse
|
227
|
Abstract
Non-Hodgkin lymphoma encompasses a diverse group of B-cell and T-cell neoplasms. Current classification is based on clinical information, histologic assessment, immunophenotypic characteristics, and molecular alterations. A wide range of genetic alterations, including large chromosomal structural rearrangements, aneuploidies, point mutations, and copy number alterations, have been reported across all types of lymphomas. Many of these are now incorporated into the World Health Organization-defined criteria for the diagnostic evaluation of patients with lymphoid proliferations and, therefore, their accurate identification is paramount for diagnosis, subclassification, and selection of treatment. In addition to their value in the diagnostic setting, many alterations that are not routinely evaluated in standard clinical practice may still define specific disease entities as they have important implications in risk stratification, as well as roles in emerging alternate therapies and disease monitoring. Because of the complexity and range of alterations, their accurate and sensitive assessment requires a careful selection of technology. Here, we discuss the most commonly used molecular techniques in current clinical practice and highlight some of the benefits and pitfalls based on the type of alteration.
Collapse
|
228
|
Conde E, Hernandez S, Benito A, Caminoa A, Garrido P, Lopez-Rios F. Screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas using the VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody. Expert Rev Mol Diagn 2021; 21:437-444. [PMID: 33899645 DOI: 10.1080/14737159.2021.1919512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The development of several ROS1 inhibitors means that the importance of accurately identifying ROS1-positive lung cancer patients has never been greater. Therefore, it is crucial that ROS1 testing assays become more standardized.Areas covered: Based on primary literature, combined with personal diagnostic and research experience, this review provide a pragmatic update on the use of the recently released VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody.Expert opinion: This assay provides high sensitivity, so it is an excellent analytical option when screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas.
Collapse
Affiliation(s)
- Esther Conde
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, CIBERONC, Madrid, Spain
| | - Susana Hernandez
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Amparo Benito
- Pathology, Ramon Y Cajal University Hospital, Madrid, Spain
| | | | - Pilar Garrido
- Medical Oncology, Ramon Y Cajal University Hospital, CIBERONC, Madrid, Spain
| | - Fernando Lopez-Rios
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, CIBERONC, Madrid, Spain
| |
Collapse
|
229
|
Targeted RNA expression profiling identifies high-grade endometrial stromal sarcoma as a clinically relevant molecular subtype of uterine sarcoma. Mod Pathol 2021; 34:1008-1016. [PMID: 33077922 DOI: 10.1038/s41379-020-00705-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
High-grade endometrial stromal sarcoma (HGESS) may harbor YWHAE-NUTM2A/B fusion, ZC3H7B-BCOR fusion, and BCOR internal tandem duplication (ITD). NTRK3 upregulation and pan-Trk expression were reported in soft tissue lesions that share similar morphology and genetic abnormalities. To confirm these findings in HGESS, differential expression analysis was performed at gene level comparing 11 HGESS with 48 other uterine sarcomas, including 9 low-grade endometrial stromal sarcomas, 23 undifferentiated uterine sarcomas, and 16 leiomyosarcomas, using targeted RNA sequencing data. Pan-Trk immunohistochemistry was performed on 35 HGESS, including 10 tumors with RNA expression data, with genotypes previously confirmed by targeted RNA sequencing, fluorescence in situ hybridization, and/or genomic PCR. Unsupervised hierarchical clustering of the top 25% of differentially expressed probes identified three molecular groups: (1) high NTRK3, FGFR3, RET, BCOR, GLI1, and PTCH1 and low ESR1 expression; (2) low NTRK3, FGFR3, RET, BCOR, GLI1, and PTCH1 and high ESR1 expression; and (3) low NTRK3, FGFR3, RET, BCOR, GLI1, PTCH1, and ESR1 expression. Among HGESS, 64% of tumors clustered in group 1, while 27% clustered in group 2. Cytoplasmic and/or nuclear pan-Trk staining of variable extent and intensity was seen in 91% of HGESS regardless of cyclin D1 and/or BCOR positivity. ER and PR expression was seen in 44% of HGESS despite ESR1 downregulation. Two patients with ER and PR positive but ESR1 downregulated stage I HGESS were treated with endocrine therapy, and both recurred at 12 and 36 months after primary resection. By RNA expression, HGESS appear homogenous and distinct from other uterine sarcomas by activation of kinases, including NTRK3, and sonic hedgehog pathway genes along with downregulation of ESR1. Most HGESS demonstrate pan-Trk staining which may serve as a diagnostic biomarker. ESR1 downregulation is seen in some HGESS that express ER and PR which raises implications in the utility of endocrine therapy in these patients.
Collapse
|
230
|
Ramani NS, Chen H, Broaddus R, Lazar AJ, Luthra R, Medeiros LJ, Patel KP, Rashid A, Routbort MJ, Stewart J, Tang Z, Bassett R, Manekia J, Barkoh BA, Dang H, Roy-Chowdhuri S. Utilization of cytology smears improves success rates of RNA-based next-generation sequencing gene fusion assays for clinically relevant predictive biomarkers. Cancer Cytopathol 2021; 129:374-382. [PMID: 33119213 PMCID: PMC12002355 DOI: 10.1002/cncy.22381] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The use of RNA-based next-generation sequencing (NGS) assays to detect gene fusions for targeted therapy has rapidly become an essential component of comprehensive molecular profiling. For cytology specimens, the cell block (CB) is most commonly used for fusion testing; however, insufficient cellularity and/or suboptimal RNA quality are often limiting factors. In the current study, the authors evaluated the factors affecting RNA fusion testing in cytology and the added value of smears in cases with a suboptimal or inadequate CB. METHODS A 12-month retrospective review was performed to identify cytology cases that were evaluated by a targeted RNA-based NGS assay. Samples were sequenced by targeted amplicon-based NGS for 51 clinically relevant genes on a proprietary platform. Preanalytic factors and NGS quality parameters were correlated with the results of RNA fusion testing. RESULTS The overall success rate of RNA fusion testing was 92%. Of the 146 cases successfully sequenced, 14% had a clinically relevant fusion detected. NGS testing success positively correlated with RNA yield (P = .03) but was independent of the tumor fraction, the tumor size, or the number of slides used for extraction. CB preparations were adequate for testing in 45% cases, but the inclusion of direct smears increased the adequacy rate to 92%. There was no significant difference in testing success rates between smears and CB preparations. CONCLUSIONS The success of RNA-based NGS fusion testing depends on the quality and quantity of RNA extracted. The use of direct smears significantly improves the adequacy of cytologic samples for RNA fusion testing for predictive biomarkers.
Collapse
Affiliation(s)
- Nisha S. Ramani
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Russell Broaddus
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexander J. Lazar
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Keyur P. Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Asif Rashid
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mark J. Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John Stewart
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Roland Bassett
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jawad Manekia
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bedia A. Barkoh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hyvan Dang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sinchita Roy-Chowdhuri
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
231
|
Li W, Li Y, Guo L, Liu Y, Yang L, Ying J. Metastatic NSCLCs With Limited Tissues: How to Effectively Identify Driver Alterations to Guide Targeted Therapy in Chinese Patients. JTO Clin Res Rep 2021; 2:100167. [PMID: 34590019 PMCID: PMC8474491 DOI: 10.1016/j.jtocrr.2021.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Molecular diagnostics of newly diagnosed patients with metastatic NSCLC (mNSCLC) with limited tissue samples often face several obstacles in routine practice using next-generation sequencing (NGS), mainly owing to insufficient tissue or DNA; thus, how to effectively identify the molecular profiling of these cases to accurately guide targeted therapy remains elusive. We evaluated whether an optimized workflow with the combined use of multiple technologies could be helpful. METHODS Tissue NGS was used as the frontline method. Amplification refractory mutation system polymerase chain reaction, immunohistochemistry, fluorescence in situ hybridization, and plasma NGS were used as supplements. RESULTS Among 208 mNSCLC cases with limited tissue (cohort 1), molecular genotyping using single-tissue NGS failed in 42 (20.2%) and actionable alterations were identified in only 112 of 208 cases (53.8%). In comparison, the optimized workflow in 1184 additional mNSCLC cases with limited tissue (cohort 2) increased the discovery rate of actionable alterations from 59.7% detected by tissue NGS to 70.4%. It was because that driver alterations were identified using amplification refractory mutation system polymerase chain reaction plus immunohistochemistry or fluorescence in situ hybridization in 53 of 78 (67.9%) tissue NGS-failed cases, and using plasma NGS in 73 of 143 (51.0%) tissue NGS-failed cases, which led to matched targeted therapies in 57 cases with clinical response. Moreover, the median turnaround time of the optimized workflow was significantly shorter than that of repeated biopsy for tissue NGS (p < 0.001). CONCLUSIONS The optimized workflow can improve mutation detection and may avoid repeated biopsy, thus allowing the timely initiation of targeted therapies for patients with newly diagnosed mNSCLC.
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
| | - Yan 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 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
| | - 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
|
232
|
Socinski MA, Pennell NA, Davies KD. MET Exon 14 Skipping Mutations in Non-Small-Cell Lung Cancer: An Overview of Biology, Clinical Outcomes, and Testing Considerations. JCO Precis Oncol 2021; 5:PO.20.00516. [PMID: 34036238 PMCID: PMC8140815 DOI: 10.1200/po.20.00516] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Nathan A. Pennell
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Kurtis D. Davies
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
233
|
Novel Preclinical Patient-Derived Lung Cancer Models Reveal Inhibition of HER3 and MTOR Signaling as Therapeutic Strategies for NRG1 Fusion-Positive Cancers. J Thorac Oncol 2021; 16:1149-1165. [PMID: 33839363 DOI: 10.1016/j.jtho.2021.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION NRG1 rearrangements produce chimeric ligands that subvert the ERBB pathway to drive tumorigenesis. A better understanding of the signaling networks that mediate transformation by NRG1 fusions is needed to inform effective therapeutic strategies. Unfortunately, this has been hampered by a paucity of patient-derived disease models that faithfully recapitulate this molecularly defined cancer subset. METHODS Patient-derived xenograft (PDX) and cell line models were established from NRG1-rearranged lung adenocarcinoma samples. Transcriptomic, proteomic, and biochemical analyses were performed to identify activated pathways. Efficacy studies were conducted to evaluate HER3- and MTOR-directed therapies. RESULTS We established a pair of PDX and cell line models of invasive mucinous lung adenocarcinoma (LUAD) (LUAD-0061AS3, SLC3A2-NRG1), representing the first reported paired in vitro and in vivo model of NRG1-driven tumors. Growth of LUAD-0061AS3 models was reduced by the anti-HER3 antibody GSK2849330. Transcriptomic profiling revealed activation of the MTOR pathway in lung tumor samples with NRG1 fusions. Phosphorylation of several MTOR effectors (S6 and 4EBP1) was higher in LUAD-0061AS3 cells compared with human bronchial epithelial cells and the breast cancer cell line MDA-MB-175-VII (DOC4-NRG1 fusion). Accordingly, LUAD-0061AS3 cells were more sensitive to MTOR inhibitors than MDA-MB-175-VII cells and targeting the MTOR pathway with rapamycin blocked growth of LUAD-0061AS3 PDX tumors in vivo. In contrast, MDA-MB-175-VII breast cancer cells had higher MAPK pathway activation and were more sensitive to MEK inhibition. CONCLUSIONS We identify the MTOR pathway as a candidate vulnerability in NRG1 fusion-positive lung adenocarcinoma that may warrant further preclinical evaluation, with the eventual goal of finding additional therapeutic options for patients in whom ERBB-directed therapy fails. Moreover, our results uncover heterogeneity in downstream oncogenic signaling among NRG1-rearranged cancers, possibly tumor type-dependent, the therapeutic significance of which requires additional investigation.
Collapse
|
234
|
Lee PC, Hendifar A, Osipov A, Cho M, Li D, Gong J. Targeting the Fibroblast Growth Factor Receptor (FGFR) in Advanced Cholangiocarcinoma: Clinical Trial Progress and Future Considerations. Cancers (Basel) 2021; 13:1706. [PMID: 33916849 PMCID: PMC8038487 DOI: 10.3390/cancers13071706] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/15/2021] [Accepted: 03/31/2021] [Indexed: 01/06/2023] Open
Abstract
Landmark molecular profiling efforts have identified multiple targetable alterations in cholangiocarcinoma. Among the molecular-driven subsets of cholangiocarcinoma, targeting the fibroblast growth factor receptor (FGFR) has shown promise and represents the first targeted therapy to be approved in treatment-refractory, advanced cholangiocarcinoma. In this review, we provide an up-to-date overview of the clinical development of FGFR inhibitors in advanced cholangiocarcinoma. We review the FGFR pathway and discuss emerging issues including resistance to FGFR inhibitors. We end with a discussion on future considerations to optimize the potential of this class of therapeutics in advanced cholangiocarcinoma.
Collapse
Affiliation(s)
- Patrick C. Lee
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.C.L.); (A.H.); (A.O.)
| | - Andrew Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.C.L.); (A.H.); (A.O.)
| | - Arsen Osipov
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.C.L.); (A.H.); (A.O.)
| | - May Cho
- Department of Medicine, UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
- UCI Health Chao Family Comprehensive Cancer Center, University of California Irvine, Orange, CA 92868, USA
| | - Daneng Li
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Jun Gong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.C.L.); (A.H.); (A.O.)
| |
Collapse
|
235
|
Suehara Y, Kohsaka S, Hayashi T, Akaike K, Kurisaki-Arakawa A, Sato S, Kobayashi E, Mizuno S, Ueno T, Morii T, Okuma T, Kurihara T, Hasegawa N, Sano K, Sasa K, Okubo T, Kim Y, Mano H, Saito T. Identification of a Novel MAN1A1-ROS1 Fusion Gene Through mRNA-based Screening for Tyrosine Kinase Gene Aberrations in a Patient with Leiomyosarcoma. Clin Orthop Relat Res 2021; 479:838-852. [PMID: 33196586 PMCID: PMC8083907 DOI: 10.1097/corr.0000000000001548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Soft tissue sarcomas are a heterogeneous group of rare malignant tumors. Advanced soft tissue sarcomas have a poor prognosis, and effective systemic therapies have not been established. Tyrosine kinases are increasingly being used as therapeutic targets for a variety of cancers and soft tissue sarcomas. Although complex karyotype sarcomas typically tend to carry more potentially actionable genetic alterations than do translocation-associated sarcomas (fusion gene sarcomas), based on our database review, we found that leiomyosarcoma and malignant peripheral nerve sheath tumors have lower frequencies of potential targets than other nontranslocation soft tissue sarcomas. We theorized that both leiomyosarcoma and malignant peripheral nerve sheath tumors might be included in any unique translocations. Furthermore, if tyrosine kinase imbalances, especially fusion genes, occur in patients with leiomyosarcomas and malignant peripheral nerve sheath tumors, tyrosine kinase inhibitors might be a drug development target for this sarcoma. In this study, we used a tyrosine kinase screening system that could detect an imbalance in mRNA between 5'- and 3'-sides in tyrosine kinase genes to identify potential novel therapeutic tyrosine kinase targets for soft tissue sarcomas. QUESTIONS/PURPOSES (1) Are there novel therapeutic tyrosine kinase targets in tumors from patients with soft tissue sarcomas that are detectable using mRNA screening focusing on imbalance expressions between the 5' and 3' end of the kinase domain? (2) Can potential targets be verified by RNA sequencing and reverse transcription PCR (RT-PCR)? (3) Will potential fusion gene(s) transform cells in in vitro assays? (4) Will tumors in mice that have an identified fusion gene respond to treatment with a therapeutic drug directed at that target? METHODS We used mRNA screening to look for novel tyrosine kinase targets that might be of therapeutic potential. Using functional assays, we verified whether the identified fusion genes would be good therapeutic candidates for soft tissue sarcomas. Additionally, using in vivo assays, we assessed whether suppressing the fusion's kinase activity has therapeutic potential. Study eligibility was based on a patient having high-grade spindle cell and nontranslocation sarcomas, including leiomyosarcoma, malignant peripheral nerve sheath tumor, and high-grade myxofibrosarcoma. Between 2015 and 2019, of the 172 patients with soft tissue sarcomas treated with surgical resection at Juntendo University Hospital, 72 patients had high-grade nontranslocation sarcomas. The analysis was primarily for leiomyosarcoma and malignant peripheral nerve sheath tumors, and there was a limitation of analysis size (reagent limitations) totaling 24 samples at the start of the study. We collected additional samples from a sample bank at the Tokyo Medical and Dental University to increase the number of sarcomas to study. Therefore, in this study, a total of 15 leiomyosarcoma samples, five malignant peripheral nerve sheath tumors samples, and four high-grade myxofibrosarcoma samples were collected to achieve the sample size of 24 patients. To identify tyrosine kinase fusion genes, we designed a NanoString-based assay (NanoString Technologies Inc, Seattle, WA, USA) to query the expression balances regarding transcripts of 90 tyrosine kinases at two points: the 5' end of the kinase domain and within the kinase domain or 3' end of the kinase domain. The tumor's RNA was hybridized to the NanoString probes and analyzed for the expression ratios of outliers from the 3' to 5' end of the kinase domain. Presumed novel fusion events in these positive tumors that were defined by NanoString-based assays were confirmed tyrosine kinase fusion genes by RNA sequencing and confirmatory RT-PCR. Functional analyses consisting of in vitro and in vivo assays were also performed to elucidate whether the identified tyrosine kinase gene fusions were associated with oncogenic abilities and drug responses. RESULTS We identified aberrant expression ratios regarding the 3' to 5' end of the kinase domain ratios in ROS1 transcripts in a leiomyosarcoma in a 90-year-old woman. A novel MAN1A1-ROS1 fusion gene was identified from her thigh tumor through RNA sequencing, which was confirmed with real-time PCR. In functional assays, MAN1A1-ROS1 rearrangement revealed strong transforming potential in 3T3 cells. Moreover, in an in vivo assay, crizotinib, a ROS1 inhibitor, markedly inhibited the growth of MAN1A1-ROS1 rearrangement-induced transformed cells in a dose-dependent manner. CONCLUSION We conducted tyrosine kinase screening to identify new therapeutic targets in soft tissue sarcomas. We found a novel MAN1A1-ROS1 fusion gene that may be a therapeutic target in patients with leiomyosarcoma. This study demonstrates that the mRNA screening system may aid in the development of useful therapeutic options for soft tissue sarcomas. CLINICAL RELEVANCE If novel tyrosine fusions such as MAN1A1-ROS1 fusion can be found in sarcomas from other patients, they could offer avenues for new molecular target therapies for sarcomas that currently do not have effective chemotherapeutic options. Therefore, the establishment of a screening system that includes both genomic and transcript analyses in the clinical setting is needed to verify our discoveries and take the developmental process of treatment to the next step.
Collapse
Affiliation(s)
- Yoshiyuki Suehara
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shinji Kohsaka
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Takuo Hayashi
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keisuke Akaike
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Aiko Kurisaki-Arakawa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shingo Sato
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Eisuke Kobayashi
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Sho Mizuno
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Toshihide Ueno
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Takeshi Morii
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Tomotake Okuma
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Taisei Kurihara
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Nobuhiko Hasegawa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Kei Sano
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keita Sasa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Taketo Okubo
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Youngji Kim
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Hiroyuki Mano
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Tsuyoshi Saito
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
236
|
Therapeutic strategies in RET gene rearranged non-small cell lung cancer. J Hematol Oncol 2021; 14:50. [PMID: 33771190 PMCID: PMC7995721 DOI: 10.1186/s13045-021-01063-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
The recent approvals by the Food and Drug Administration several tumor-agnostic drugs have resulted in a paradigm shift in cancer treatment from an organ/histology-specific strategy to biomarker-guided approaches. RET gene fusions are oncogenic drivers in multiple tumor types and are known to occur in 1-2% of non-squamous NSCLC patients. RET gene fusions give rise to chimeric, cytosolic proteins with constitutively active RET kinase domain. Standard therapeutic regimens provide limited benefit for NSCLC patients with RET fusion-positive tumors, and the outcomes with immunotherapy in the these patients are generally poor. Selpercatinib (LOXO-292) and pralsetinib (BLU-667) are potent and selective inhibitors that target RET alterations, including fusions and mutations, irrespective of the tissue of origin. Recently, the results from the LIBRETTO-001 and ARROW clinical trials demonstrated significant clinical benefits with selpercatinib and pralsetinib respectively, in NSCLC patients with RET gene fusions, with tolerable toxicity profiles. These studies also demonstrated that these RET-TKIs crossed the blood brain barrier with significant activity. As has been observed with other TKIs, the emergence of acquired resistance may limit long-term efficacy of these agents. Therefore, understanding the mechanisms of resistance is necessary for the development of strategies to overcome them.
Collapse
|
237
|
Tan AC, Loh TJ, Kwang XL, Tan GS, Lim KH, Tan DSW. Novel Therapies for Metastatic Non-Small Cell Lung Cancer with MET Exon 14 Alterations: A Spotlight on Capmatinib. LUNG CANCER-TARGETS AND THERAPY 2021; 12:11-20. [PMID: 33776501 PMCID: PMC7987308 DOI: 10.2147/lctt.s263610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
MET exon 14 (METex14) alterations are now an established therapeutically tractable target in non-small cell lung cancer (NSCLC). Recently reported trials of several MET tyrosine kinase inhibitors (TKI) in this patient population have demonstrated promising efficacy data in both the treatment naïve and pre-treated settings and have led to regulatory approvals. This review will focus on practical diagnostic considerations for METex14 alterations, the trial evidence for capmatinib in this molecular subset including dosing and toxicity management, and the future therapeutic landscape of METex14 altered NSCLC.
Collapse
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Tracy J Loh
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Xue Lin Kwang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Gek San Tan
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| |
Collapse
|
238
|
Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, Bruno DS, Chang JY, Chirieac LR, D'Amico TA, Dilling TJ, Dowell J, Gettinger S, Gubens MA, Hegde A, Hennon M, Lackner RP, Lanuti M, Leal TA, Lin J, Loo BW, Lovly CM, Martins RG, Massarelli E, Morgensztern D, Ng T, Otterson GA, Patel SP, Riely GJ, Schild SE, Shapiro TA, Singh AP, Stevenson J, Tam A, Yanagawa J, Yang SC, Gregory KM, Hughes M. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 2.2021. J Natl Compr Canc Netw 2021; 19:254-266. [PMID: 33668021 DOI: 10.6004/jnccn.2021.0013] [Citation(s) in RCA: 632] [Impact Index Per Article: 158.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Non-Small Cell Lung Cancer (NSCLC) address all aspects of management for NSCLC. These NCCN Guidelines Insights focus on recent updates to the NCCN Guidelines regarding targeted therapies, immunotherapies, and their respective biomarkers.
Collapse
Affiliation(s)
| | - Douglas E Wood
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | | | - Ankit Bharat
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Debora S Bruno
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - Joe Y Chang
- The University of Texas MD Anderson Cancer Center
| | | | | | | | | | | | | | | | | | | | | | | | - Jules Lin
- University of Michigan Rogel Cancer Center
| | | | | | - Renato G Martins
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Daniel Morgensztern
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Thomas Ng
- The University of Tennessee Health Science Center
| | - Gregory A Otterson
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | | | | | | | - Aditi P Singh
- Abramson Cancer Center at the University of Pennsylvania
| | - James Stevenson
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - Alda Tam
- The University of Texas MD Anderson Cancer Center
| | | | - Stephen C Yang
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | |
Collapse
|
239
|
Song Z, Lu C, Xu CW, Zheng Z. Noncanonical Gene Fusions Detected at the DNA Level Necessitate Orthogonal Diagnosis Methods Before Targeted Therapy. J Thorac Oncol 2021; 16:344-348. [PMID: 33641715 DOI: 10.1016/j.jtho.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Zhengbo Song
- Department of Clinical Trial, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China; Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Chenyu Lu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China; Biotechnology and Health Centre, City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
| | - Chun-Wei Xu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Zongli Zheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China; Biotechnology and Health Centre, City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China; Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong, People's Republic of China.
| |
Collapse
|
240
|
Krook MA, Reeser JW, Ernst G, Barker H, Wilberding M, Li G, Chen HZ, Roychowdhury S. Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br J Cancer 2021; 124:880-892. [PMID: 33268819 PMCID: PMC7921129 DOI: 10.1038/s41416-020-01157-0] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/06/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are aberrantly activated through single-nucleotide variants, gene fusions and copy number amplifications in 5-10% of all human cancers, although this frequency increases to 10-30% in urothelial carcinoma and intrahepatic cholangiocarcinoma. We begin this review by highlighting the diversity of FGFR genomic alterations identified in human cancers and the current challenges associated with the development of clinical-grade molecular diagnostic tests to accurately detect these alterations in the tissue and blood of patients. The past decade has seen significant advancements in the development of FGFR-targeted therapies, which include selective, non-selective and covalent small-molecule inhibitors, as well as monoclonal antibodies against the receptors. We describe the expanding landscape of anti-FGFR therapies that are being assessed in early phase and randomised controlled clinical trials, such as erdafitinib and pemigatinib, which are approved by the Food and Drug Administration for the treatment of FGFR3-mutated urothelial carcinoma and FGFR2-fusion cholangiocarcinoma, respectively. However, despite initial sensitivity to FGFR inhibition, acquired drug resistance leading to cancer progression develops in most patients. This phenomenon underscores the need to clearly delineate tumour-intrinsic and tumour-extrinsic mechanisms of resistance to facilitate the development of second-generation FGFR inhibitors and novel treatment strategies beyond progression on targeted therapy.
Collapse
Affiliation(s)
- Melanie A Krook
- Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Julie W Reeser
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabrielle Ernst
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Hannah Barker
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Max Wilberding
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gary Li
- QED Therapeutics Inc., San Francisco, CA, USA
| | - Hui-Zi Chen
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Sameek Roychowdhury
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
241
|
Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD, Oxnard GR. Circulating tumor DNA in advanced solid tumors: Clinical relevance and future directions. CA Cancer J Clin 2021; 71:176-190. [PMID: 33165928 DOI: 10.3322/caac.21650] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
The application of genomic profiling assays using plasma circulating tumor DNA (ctDNA) is rapidly evolving in the management of patients with advanced solid tumors. Diverse plasma ctDNA technologies in both commercial and academic laboratories are in routine or emerging use. The increasing integration of such testing to inform treatment decision making by oncology clinicians has complexities and challenges but holds significant potential to substantially improve patient outcomes. In this review, the authors discuss the current role of plasma ctDNA assays in oncology care and provide an overview of ongoing research that may inform real-world clinical applications in the near future.
Collapse
Affiliation(s)
- Michael L Cheng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Eirini Pectasides
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Heather A Parsons
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Atish D Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey R Oxnard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
242
|
Belli C, Penault-Llorca F, Ladanyi M, Normanno N, Scoazec JY, Lacroix L, Reis-Filho JS, Subbiah V, Gainor JF, Endris V, Repetto M, Drilon A, Scarpa A, André F, Douillard JY, Curigliano G. ESMO recommendations on the standard methods to detect RET fusions and mutations in daily practice and clinical research. Ann Oncol 2021; 32:337-350. [PMID: 33455880 DOI: 10.1016/j.annonc.2020.11.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Aberrant activation of RET is a critical driver of growth and proliferation in diverse solid tumours. Multikinase inhibitors (MKIs) showing anti-RET activities have been tested in RET-altered tumours with variable results. The low target specificity with consequent increase in side-effects and off-target toxicities resulting in dose reduction and drug discontinuation are some of the major issues with MKIs. To overcome these issues, new selective RET inhibitors such as pralsetinib (BLU-667) and selpercatinib (LOXO-292) have been developed in clinical trials, with selpercatinib recently approved by the Food and Drug Administration (FDA). The results of these trials showed marked and durable antitumour activity and manageable toxicity profiles in patients with RET-altered tumours. The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group (TR and PM WG) launched a collaborative project to review the available methods for the detection of RET gene alterations, their potential applications and strategies for the implementation of a rational approach for the detection of RET fusion genes and mutations in human malignancies. We present here recommendations for the routine clinical detection of targetable RET rearrangements and mutations.
Collapse
Affiliation(s)
- C Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - F Penault-Llorca
- University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Department of BioPathology, Clermont-Ferrand, France
| | - M Ladanyi
- Department of Pathology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - N Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - J-Y Scoazec
- AMMICa, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy, Villejuif, France; Department of Pathology and Translational Research, Gustave Roussy Cancer Centre, Villejuif, France
| | - L Lacroix
- Translational Research Laboratory and Biobank, Gustave Roussy, Villejuif, France; Inserm U981, Gustave Roussy, Villejuif, France; Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - J S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - V Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J F Gainor
- Massachusetts General Hospital, Boston, USA
| | - V Endris
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - M Repetto
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - A Drilon
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - A Scarpa
- ARC-Net Research Centre and Department of Diagnostics and Public Health - Section of Pathology, University of Verona, Verona, Italy
| | - F André
- Gustave Roussy Cancer Center, Villejuif, France
| | - J-Y Douillard
- Scientific and Medical Division, European Society for Medical Oncology, Lugano, Switzerland
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
243
|
Li J, Zhang B, Zhang Y, Xu F, Zhang Z, Shao L, Yan C, Ulivi P, Denis MG, Christopoulos P, Thomas de Montpréville V, Bernicker EH, van der Wekken AJ, Wang C, Yue D. Concomitant mutation status of ALK-rearranged non-small cell lung cancers and its prognostic impact on patients treated with crizotinib. Transl Lung Cancer Res 2021; 10:1525-1535. [PMID: 33889527 PMCID: PMC8044492 DOI: 10.21037/tlcr-21-160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background In non-small cell lung cancer (NSCLC), anaplastic lymphoma kinase (ALK) rearrangement characterizes a subgroup of patients who show sensitivity to ALK tyrosine kinase inhibitors (TKIs). However, the prognoses of these patients are heterogeneous. A better understanding of the genomic alterations occurring in these tumors could explain the prognostic heterogeneity observed in these patients. Methods We retrospectively analyzed 96 patients with NSCLC with ALK detected by immunohistochemical staining (VENTANA anti-ALK(D5F3) Rabbit Monoclonal Primary Antibody). Cancer tissues were subjected to next-generation sequencing using a panel of 520 cancer-related genes. The genomic landscape, distribution of ALK fusion variants, and clinicopathological characteristics of the patients were evaluated. The correlations of genomic alterations with clinical outcomes were also assessed. Results Among the 96 patients with immunohistochemically identified ALK fusions, 80 (83%) were confirmed by next-generation sequencing. TP53 mutation was the most commonly co-occurring mutation with ALK rearrangement. Concomitant driver mutations [2 Kirsten rat sarcoma viral oncogene homolog (KRAS) G12, 1 epidermal growth factor receptor (EGFR) 19del, and 1 MET exon 14 skipping] were also observed in 4 adenocarcinomas. Echinoderm microtubule associated protein-like 4 (EML4)-ALK fusions were identified in 95% of ALK-rearranged patients, with 16.2% of them also harboring additional non-EML4-ALK fusions. Nineteen non-EML4 translocation partners were also discovered, including 10 novel ones. Survival analyses revealed that patients concurrently harboring PIK3R2 alterations showed a trend toward shorter progression-free survival (6 vs. 13 months, P=0.064) and significantly shorter overall survival (11 vs. 32 months, P=0.004) than did PIK3R2-wild-type patients. Patients with concomitant alterations in PI3K the signaling pathway also had a shorter median overall survival than those without such alterations (23 vs. 32 months, P=0.014), whereas progression-free survival did not differ significantly. Conclusions The spectrum of ALK-fusion variants and the landscape of concomitant genomic alterations were delineated in 96 NSCLC patients. Our study also demonstrated the prognostic value of concomitant alterations in crizotinib-treated patients, which could facilitate improved stratification of ALK-rearranged NSCLC patients in the selection of candidates who could optimally benefit from therapy.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Bin Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Yu Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Feng Xu
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Lin Shao
- Burning Rock Biotech, Beijing, China
| | | | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Marc G Denis
- Department of Biochemistry and INSERM U1232, Nantes University Hospital, Nantes Cedex, France
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases at the Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | | | | | | | - Changli Wang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Dongsheng Yue
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| |
Collapse
|
244
|
Yang SR, Aypar U, Rosen EY, Mata DA, Benayed R, Mullaney K, Jayakumaran G, Zhang Y, Frosina D, Drilon A, Ladanyi M, Jungbluth AA, Rekhtman N, Hechtman JF. A Performance Comparison of Commonly Used Assays to Detect RET Fusions. Clin Cancer Res 2021; 27:1316-1328. [PMID: 33272981 PMCID: PMC8285056 DOI: 10.1158/1078-0432.ccr-20-3208] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/23/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023]
Abstract
PURPOSE Selpercatinib and pralsetinib induce deep and durable responses in patients with advanced RET fusion-positive lung and thyroid cancer. RET fusion testing strategies with rapid and reliable results are critical given recent FDA approval. Here, we assess various clinical assays in a large pan-cancer cohort. EXPERIMENTAL DESIGN Tumors underwent DNA-based next-generation sequencing (NGS) with reflex to RNA-based NGS if no mitogenic driver or if a RET structural variant of unknown significance (SVUS) were present. Canonical DNA-level RET fusions and RNA-confirmed RET fusions were considered true fusions. Break-apart FISH and IHC performance were assessed in subgroups. RESULTS A total of 171 of 41,869 patients with DNA NGS harbored RET structural variants, including 139 canonical fusions and 32 SVUS. Twelve of 32 (37.5%) SVUS were transcribed into RNA-level fusions, resulting in 151 oncogenic RET fusions. The most common RET fusion-positive tumor types were lung (65.6%) and thyroid (23.2%). The most common partners were KIF5B (45%), CCDC6 (29.1%), and NCOA4 (13.3%). DNA NGS showed 100% (46/46) sensitivity and 99.6% (4,459/4,479) specificity. FISH showed 91.7% (44/48) sensitivity, with lower sensitivity for NCOA4-RET (66.7%, 8/12). A total of 87.5% (7/8) of RET SVUS negative for RNA-level fusions demonstrated rearrangement by FISH. The sensitivity of IHC varied by fusion partner: KIF5B sensitivity was highest (100%, 31/31), followed by CCDC6 (88.9%, 16/18) and NCOA4 (50%, 6/12). Specificity of RET IHC was 82% (73/89). CONCLUSIONS Although DNA sequencing has high sensitivity and specificity, RNA sequencing of RET SVUS is necessary. Both FISH and IHC demonstrated lower sensitivity for NCOA4-RET fusions.
Collapse
Affiliation(s)
- Soo-Ryum Yang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Umut Aypar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ezra Y Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Douglas A Mata
- 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
| | - Kerry Mullaney
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gowtham Jayakumaran
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Denise Frosina
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- 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
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
245
|
Heydt C, Wölwer CB, Velazquez Camacho O, Wagener-Ryczek S, Pappesch R, Siemanowski J, Rehker J, Haller F, Agaimy A, Worm K, Herold T, Pfarr N, Weichert W, Kirchner T, Jung A, Kumbrink J, Goering W, Esposito I, Buettner R, Hillmer AM, Merkelbach-Bruse S. Detection of gene fusions using targeted next-generation sequencing: a comparative evaluation. BMC Med Genomics 2021; 14:62. [PMID: 33639937 PMCID: PMC7912891 DOI: 10.1186/s12920-021-00909-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 02/17/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Gene fusions represent promising targets for cancer therapy in lung cancer. Reliable detection of multiple gene fusions is therefore essential. METHODS Five commercially available parallel sequencing assays were evaluated for their ability to detect gene fusions in eight cell lines and 18 FFPE tissue samples carrying a variety of known gene fusions. Four RNA-based assays and one DNA-based assay were compared; two were hybrid capture-based, TruSight Tumor 170 Assay (Illumina) and SureSelect XT HS Custom Panel (Agilent), and three were amplicon-based, Archer FusionPlex Lung Panel (ArcherDX), QIAseq RNAscan Custom Panel (Qiagen) and Oncomine Focus Assay (Thermo Fisher Scientific). RESULTS The Illumina assay detected all tested fusions and showed the smallest number of false positive results. Both, the ArcherDX and Qiagen panels missed only one fusion event. Among the RNA-based assays, the Qiagen panel had the highest number of false positive events. The Oncomine Focus Assay (Thermo Fisher Scientific) was the least adequate assay for our purposes, seven fusions were not covered by the assay and two fusions were classified as uncertain. The DNA-based SureSelect XT HS Custom Panel (Agilent) missed three fusions and nine fusions were only called by one software version. Additionally, many false positive fusions were observed. CONCLUSIONS In summary, especially RNA-based parallel sequencing approaches are potent tools for reliable detection of targetable gene fusions in clinical diagnostics.
Collapse
Affiliation(s)
- Carina Heydt
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Christina B Wölwer
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Oscar Velazquez Camacho
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Svenja Wagener-Ryczek
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Roberto Pappesch
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Janna Siemanowski
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Jan Rehker
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Florian Haller
- Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Karl Worm
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Herold
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nicole Pfarr
- Institute of Pathology, Technical University Munich (TUM), Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich (TUM), Munich, Germany
| | | | - Andreas Jung
- Institute of Pathology, LMU Munich, Munich, Germany
| | | | - Wolfgang Goering
- Institute of Pathology, Medical Faculty, Heinrich-Heine-University and University Hospital Duesseldorf, Düesseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty, Heinrich-Heine-University and University Hospital Duesseldorf, Düesseldorf, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Axel M Hillmer
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
246
|
Siemanowski J, Heydt C, Merkelbach-Bruse S. Predictive molecular pathology of lung cancer in Germany with focus on gene fusion testing: Methods and quality assurance. Cancer Cytopathol 2021; 128:611-621. [PMID: 32885916 DOI: 10.1002/cncy.22293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
Predictive molecular testing has become an important part of the diagnosis of any patient with lung cancer. Using reliable methods to ensure timely and accurate results is inevitable for guiding treatment decisions. In the past few years, parallel sequencing has been established for mutation testing, and its use is currently broadened for the detection of other genetic alterations, such as gene fusion and copy number variations. In addition, conventional methods such as immunohistochemistry and in situ hybridization are still being used, either for formalin-fixed, paraffin-embedded tissue or for cytological specimens. For the development and broad implementation of such complex technologies, interdisciplinary and regional networks are needed. The Network Genomic Medicine (NGM) has served as a model of centralized testing and decentralized treatment of patients and incorporates all German comprehensive cancer centers. Internal quality control, laboratory accreditation, and participation in external quality assessment is mandatory for the delivery of reliable results. Here, we provide a summary of current technologies used to identify patients who have lung cancer with gene fusions, briefly describe the structures of NGM and the national NGM (nNGM), and provide recommendations for quality assurance.
Collapse
Affiliation(s)
- Janna Siemanowski
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | |
Collapse
|
247
|
König D, Savic Prince S, Rothschild SI. Targeted Therapy in Advanced and Metastatic Non-Small Cell Lung Cancer. An Update on Treatment of the Most Important Actionable Oncogenic Driver Alterations. Cancers (Basel) 2021; 13:804. [PMID: 33671873 PMCID: PMC7918961 DOI: 10.3390/cancers13040804] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
Due to groundbreaking developments and continuous progress, the treatment of advanced and metastatic non-small cell lung cancer (NSCLC) has become an exciting, but increasingly challenging task. This applies, in particular, to the subgroup of NSCLC with oncogenic driver alterations. While the treatment of epidermal growth factor receptor (EGFR)-mutated and anaplastic lymphoma kinase (ALK)-rearranged NSCLC with various tyrosine kinase inhibitors (TKIs) is well-established, new targets have been identified in the last few years and new TKIs introduced in clinical practice. Even for KRAS mutations, considered for a long time as an "un-targetable" alteration, promising new drugs are emerging. The detection and in-depth molecular analysis of resistance mechanisms has further fueled the development of new therapeutic strategies. The objective of this review is to give a comprehensive overview on the current landscape of targetable oncogenic alterations in NSCLC.
Collapse
Affiliation(s)
- David König
- Department of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland;
- Comprehensive Cancer Center, University Hospital Basel, 4031 Basel, Switzerland;
| | - Spasenija Savic Prince
- Comprehensive Cancer Center, University Hospital Basel, 4031 Basel, Switzerland;
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland
| | - Sacha I. Rothschild
- Department of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland;
- Comprehensive Cancer Center, University Hospital Basel, 4031 Basel, Switzerland;
| |
Collapse
|
248
|
Prospective pan-cancer germline testing using MSK-IMPACT informs clinical translation in 751 patients with pediatric solid tumors. ACTA ACUST UNITED AC 2021; 2:357-365. [PMID: 34308366 DOI: 10.1038/s43018-021-00172-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The spectrum of germline predisposition in pediatric cancer continues to be realized. Here we report 751 solid tumor patients who underwent prospective matched tumor-normal DNA sequencing and downstream clinical use (clinicaltrials.gov NCT01775072). Germline pathogenic and likely pathogenic (P/LP) variants were reported. One or more P/LP variants were found in 18% (138/751) of individuals when including variants in low, moderate, and high penetrance dominant or recessive genes, or 13% (99/751) in moderate and high penetrance dominant genes. 34% of high or moderate penetrance variants were unexpected based on the patient's diagnosis and previous history. 76% of patients with positive results completed a clinical genetics visit, and 21% had at least one relative undergo cascade testing as a result of this testing. Clinical actionability additionally included screening, risk reduction in relatives, reproductive use, and use of targeted therapies. Germline testing should be considered for all children with cancer.
Collapse
|
249
|
Kato S. Tumour-Agnostic Therapy for Pancreatic Cancer and Biliary Tract Cancer. Diagnostics (Basel) 2021; 11:252. [PMID: 33562094 PMCID: PMC7914923 DOI: 10.3390/diagnostics11020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022] Open
Abstract
The prognosis of patients with solid tumours has remarkably improved with the development of molecular-targeted drugs and immune checkpoint inhibitors. However, the improvements in the prognosis of pancreatic cancer and biliary tract cancer is delayed compared to other carcinomas, and the 5-year survival rates of distal-stage disease are approximately 10 and 20%, respectively. However, a comprehensive analysis of tumour cells using The Cancer Genome Atlas (TCGA) project has led to the identification of various driver mutations. Evidently, few mutations exist across organs, and basket trials targeting driver mutations regardless of the primary organ are being actively conducted. Such basket trials not only focus on the gate keeper-type oncogene mutations, such as HER2 and BRAF, but also focus on the caretaker-type tumour suppressor genes, such as BRCA1/2, mismatch repair-related genes, which cause hereditary cancer syndrome. As oncogene panel testing is a vital approach in routine practice, clinicians should devise a strategy for improved understanding of the cancer genome. Here, the gene mutation profiles of pancreatic cancer and biliary tract cancer have been outlined and the current status of tumour-agnostic therapy in these cancers has been reported.
Collapse
Affiliation(s)
- Shunsuke Kato
- Department of Clinical Oncology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
250
|
Stenzinger A, van Tilburg CM, Tabatabai G, Länger F, Graf N, Griesinger F, Heukamp LC, Hummel M, Klingebiel T, Hettmer S, Vokuhl C, Merkelbach-Bruse S, Overkamp F, Reichardt P, Scheer M, Weichert W, Westphalen CB, Bokemeyer C, Ivanyi P, Loges S, Schirmacher P, Wörmann B, Bielack S, Seufferlein TTW. [Diagnosis and therapy of tumors with NTRK gene fusion]. DER PATHOLOGE 2021; 42:103-115. [PMID: 33258061 PMCID: PMC7858552 DOI: 10.1007/s00292-020-00864-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
NTRK-Genfusionen sind seltene genetische Alterationen, die tumorentitätenübergreifend vorkommen können. Während sie in den meisten soliden Tumoren nur sehr niederfrequent vorkommen, lassen sie sich in bestimmten Tumoren wie dem infantilen Fibrosarkom, dem kongenitalen mesoblastischen Nephrom und dem sekretorischen Mamma- oder Speicheldrüsenkarzinom jedoch häufig nachweisen. NTRK-Genfusionen bzw. TRK-Fusionsproteine gelten als starke onkogene Treiber. Bei Nachweis von NTRK-Genfusionen können TRK-Inhibitoren unabhängig von der Tumorentität eingesetzt werden. Vertreter sind Entrectinib und Larotrectinib. Bislang ist nur Larotrectinib in der Europäischen Union zugelassen. Für beide wurden Wirksamkeit und Verträglichkeit in Phase-I- und Phase-II-Studien gezeigt. Die Seltenheit der TRK-Fusionstumoren stellt diagnostische und klinische Prozesse vor große Herausforderungen: Einerseits sollen alle Patienten mit TRK-Fusionstumoren identifiziert werden, andererseits sind epidemiologische und histologische Aspekte sowie Ressourcen zu berücksichtigen. Basierend auf diesen Punkten möchten wir einen Diagnosealgorithmus für TRK-Fusionstumoren vorschlagen, außerdem stellen wir aktuelle Daten zu den TRK-Inhibitoren vor.
Collapse
Affiliation(s)
- Albrecht Stenzinger
- Allgemeine Pathologie und pathologische Anatomie, Pathologisches Institut, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland.
| | - Cornelis M van Tilburg
- Hopp-Kindertumorzentrum Heidelberg (KiTZ), Deutsches Krebsforschungszentrum (DKFZ), Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Ghazaleh Tabatabai
- Abteilung Neurologie mit interdisziplinärem Schwerpunkt Neuroonkologie, Universitätsklinikum Tübingen und Hertie-Institut für Klinische Hirnforschung, Eberhard Karls Universität Tübingen, Tübingen, Deutschland
| | - Florian Länger
- Institut für Pathologie, Medizinische Hochschule Hannover, Hannover, Deutschland
| | - Norbert Graf
- Klinik für Pädiatrische Onkologie und Hämatologie, Universitätsklinikum des Saarlandes, Medizinische Fakultät, Universität des Saarlandes, Homburg, Deutschland
| | - Frank Griesinger
- Klinik für Hämatologie und Onkologie, Universitätsklinik für Innere Medizin - Onkologie, Pius-Hospital Oldenburg, Oldenburg, Deutschland
| | | | - Michael Hummel
- Institut für Pathologie (CCM), Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Thomas Klingebiel
- Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Frankfurt, Frankfurt, Deutschland
| | - Simone Hettmer
- Klinik für Pädiatrische Hämatologie und Onkologie, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Freiburg, Freiburg, Deutschland
| | - Christian Vokuhl
- Sektion Kinderpathologie, Institut für Pathologie, Universitätsklinikum Bonn, Bonn, Deutschland
| | - Sabine Merkelbach-Bruse
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Uniklinik Köln, Köln, Deutschland
| | | | - Peter Reichardt
- Onkologie und Palliativmedizin, Helios Klinikum Berlin-Buch, Berlin, Deutschland
| | - Monika Scheer
- Pädiatrie 5 - Onkologie, Hämatologie und Immunologie, Zentrum für Kinder‑, Jugend- und Frauenmedizin - Olgahospital, Stuttgart Cancer Center, Klinikum Stuttgart, Stuttgart, Deutschland
| | - Wilko Weichert
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - C Benedikt Westphalen
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Ludwig-Maximilians-Universität München, München, Deutschland
| | - Carsten Bokemeyer
- Zentrum für Onkologie, II. Medizinische Klinik und Poliklinik (Onkologie, Hämatologie, Knochenmarktransplantation mit Abteilung für Pneumologie), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
| | - Philipp Ivanyi
- Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Medizinische Hochschule Hannover, Hannover, Deutschland
| | - Sonja Loges
- Zentrum für Onkologie, II. Medizinische Klinik und Poliklinik (Onkologie, Hämatologie, Knochenmarktransplantation mit Abteilung für Pneumologie), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland.,Zentrum für experimentelle Medizin, Institut für Tumorbiologie, Universitätsklinikum Hamburg- Eppendorf, Hamburg, Deutschland.,Abteilung für Personalisierte Medizinische Onkologie, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Deutschland.,Universitätsklinikum Mannheim, Mannheim, Deutschland
| | - Peter Schirmacher
- Allgemeine Pathologie und pathologische Anatomie, Pathologisches Institut, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland
| | - Bernhard Wörmann
- Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie (CVK), Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Stefan Bielack
- Pädiatrie 5 - Onkologie, Hämatologie und Immunologie, Zentrum für Kinder‑, Jugend- und Frauenmedizin - Olgahospital, Stuttgart Cancer Center, Klinikum Stuttgart, Stuttgart, Deutschland
| | | |
Collapse
|