801
|
Pederzoli F, Bandini M, Marandino L, Ali SM, Madison R, Chung J, Ross JS, Necchi A. Targetable gene fusions and aberrations in genitourinary oncology. Nat Rev Urol 2020; 17:613-625. [PMID: 33046892 DOI: 10.1038/s41585-020-00379-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Gene fusions result from either structural chromosomal rearrangement or aberrations caused by splicing or transcriptional readthrough. The precise and distinctive presence of fusion genes in neoplastic tissues and their involvement in multiple pathways central to cancer development, growth and survival make them promising targets for personalized therapy. In genitourinary malignancies, rearrangements involving the E26 transformation-specific family of transcription factors have emerged as very frequent alterations in prostate cancer, especially the TMPRSS2-ERG fusion. In renal malignancies, Xp11 and t(6;11) translocations are hallmarks of a distinct pathological group of tumours described as microphthalmia-associated transcription factor family translocation-associated renal cell carcinomas. Novel druggable fusion events have been recognized in genitourinary malignancies, leading to the activation of several clinical trials. For instance, ALK-rearranged renal cell carcinomas have shown responses to alectinib and crizotinib. Erdafitinib has been tested for the treatment of FGFR-rearranged bladder cancer. Other anti-fibroblast growth factor receptor 3 (FGFR3) compounds are showing promising results in the treatment of bladder cancer, including infigratinib and pemigatinib, and all are currently in clinical trials.
Collapse
Affiliation(s)
- Filippo Pederzoli
- Urological Research Institute (URI), Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy.
| | - Marco Bandini
- Urological Research Institute (URI), Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Laura Marandino
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Siraj M Ali
- Foundation Medicine Inc., Cambridge, MA, USA
| | | | - Jon Chung
- Foundation Medicine Inc., Cambridge, MA, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, MA, USA.,Upstate Medical University, Syracuse, NY, USA
| | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
802
|
Yang S, Li L, Chen D, Xiao M, Xian L. EPS15-NTRK1: a novel NTRK1 oncogenic fusion in patient with lung adenocarcinoma. J Cancer Res Clin Oncol 2020; 146:3389-3392. [PMID: 33037466 DOI: 10.1007/s00432-020-03416-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/28/2020] [Indexed: 01/18/2023]
Affiliation(s)
- Shengzhuang Yang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Lijian Li
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, 210002, China
| | - Dongsheng Chen
- 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
| | - Lei Xian
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
803
|
Qian Y, Gong Y, Fan Z, Luo G, Huang Q, Deng S, Cheng H, Jin K, Ni Q, Yu X, Liu C. Molecular alterations and targeted therapy in pancreatic ductal adenocarcinoma. J Hematol Oncol 2020; 13:130. [PMID: 33008426 PMCID: PMC7532113 DOI: 10.1186/s13045-020-00958-3] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignancy characterized by a poor prognosis and high mortality rate. Genetic mutations and altered molecular pathways serve as targets in precise therapy. Using next-generation sequencing (NGS), these aberrant alterations can be identified and used to develop strategies that will selectively kill cancerous cells in patients with PDAC. The realization of targeted therapies in patients with PDAC may be summarized by three approaches. First, because oncogenes play a pivotal role in tumorigenesis, inhibition of dysregulated oncogenes is a promising method (Table 3). Numerous researchers are developing strategies to target oncogenes, such as KRAS, NRG1, and NTRK and related molecules, although most of the results are unsatisfactory. Accordingly, emerging strategies are being developed to target these oncogenes, including simultaneously inhibiting multiple molecules or pathways, modification of mutant residues by small molecules, and RNA interference. Second, researchers have attempted to reactivate inactivated tumour suppressors or modulate related molecules. TP53, CDKN2A and SMAD4 are three major tumour suppressors involved in PDAC. Advances have been achieved in clinical and preclinical trials of therapies targeting these three genes, and further investigations are warranted. The TGF-β-SMAD4 signalling pathway plays a dual role in PDAC tumorigenesis and participates in mediating tumour-stroma crosstalk and modulating the tumour microenvironment (TME); thus, molecular subtyping of pancreatic cancer according to the SMAD4 mutation status may be a promising precision oncology technique. Finally, genes such as KDM6A and BRCA have vital roles in maintaining the structural stability and physiological functions of normal chromosomes and are deficient in some patients with PDAC, thus serving as potential targets for correcting these deficiencies and precisely killing these aberrant tumour cells. Recent clinical trials, such as the POLO (Pancreas Cancer Olaparib Ongoing) trial, have reported encouraging outcomes. In addition to genetic event-guided treatment, immunotherapies such as chimeric antigen receptor T cells (CAR-T), antibody-drug conjugates, and immune checkpoint inhibitors also exhibit the potential to target tumours precisely, although the clinical value of immunotherapies as treatments for PDAC is still limited. In this review, we focus on recent preclinical and clinical advances in therapies targeting aberrant genes and pathways and predict the future trend of precision oncology for PDAC.
Collapse
Affiliation(s)
- Yunzhen Qian
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yitao Gong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Zhiyao Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qiuyi Huang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shengming Deng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, NO.270 DongAn Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
804
|
Design, synthesis, and Structure–Activity Relationships (SAR) of 3-vinylindazole derivatives as new selective tropomyosin receptor kinases (Trk) inhibitors. Eur J Med Chem 2020; 203:112552. [DOI: 10.1016/j.ejmech.2020.112552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/07/2020] [Accepted: 06/07/2020] [Indexed: 11/20/2022]
|
805
|
Cocco E, Lee JE, Kannan S, Schram AM, Won HH, Shifman S, Kulick A, Baldino L, Toska E, Arruabarrena-Aristorena A, Kittane S, Wu F, Cai Y, Arena S, Mussolin B, Kannan R, Vasan N, Gorelick AN, Berger MF, Novoplansky O, Jagadeeshan S, Liao Y, Rix U, Misale S, Taylor BS, Bardelli A, Hechtman JF, Hyman DM, Elkabets M, de Stanchina E, Verma CS, Ventura A, Drilon A, Scaltriti M. TRK xDFG Mutations Trigger a Sensitivity Switch from Type I to II Kinase Inhibitors. Cancer Discov 2020; 11:126-141. [PMID: 33004339 DOI: 10.1158/2159-8290.cd-20-0571] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/26/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
On-target resistance to next-generation TRK inhibitors in TRK fusion-positive cancers is largely uncharacterized. In patients with these tumors, we found that TRK xDFG mutations confer resistance to type I next-generation TRK inhibitors designed to maintain potency against several kinase domain mutations. Computational modeling and biochemical assays showed that TRKAG667 and TRKCG696 xDFG substitutions reduce drug binding by generating steric hindrance. Concurrently, these mutations stabilize the inactive (DFG-out) conformations of the kinases, thus sensitizing these kinases to type II TRK inhibitors. Consistently, type II inhibitors impede the growth and TRK-mediated signaling of xDFG-mutant isogenic and patient-derived models. Collectively, these data demonstrate that adaptive conformational resistance can be abrogated by shifting kinase engagement modes. Given the prior identification of paralogous xDFG resistance mutations in other oncogene-addicted cancers, these findings provide insights into rational type II drug design by leveraging inhibitor class affinity switching to address recalcitrant resistant alterations. SIGNIFICANCE: In TRK fusion-positive cancers, TRK xDFG substitutions represent a shared liability for type I TRK inhibitors. In contrast, they represent a potential biomarker of type II TRK inhibitor activity. As all currently available type II agents are multikinase inhibitors, rational drug design should focus on selective type II inhibitor creation.This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Emiliano Cocco
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ji Eun Lee
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Helen H Won
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sophie Shifman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amanda Kulick
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura Baldino
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eneda Toska
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Srushti Kittane
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fan Wu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yanyan Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sabrina Arena
- Department of Oncology, University of Torino, Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | | | - Ram Kannan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neil Vasan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander N Gorelick
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ofra Novoplansky
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yi Liao
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida
| | - Uwe Rix
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida
| | - Sandra Misale
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alberto Bardelli
- Department of Oncology, University of Torino, Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chandra S Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - Andrea Ventura
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
806
|
Lamberti G, Andrini E, Sisi M, Rizzo A, Parisi C, Di Federico A, Gelsomino F, Ardizzoni A. Beyond EGFR, ALK and ROS1: Current evidence and future perspectives on newly targetable oncogenic drivers in lung adenocarcinoma. Crit Rev Oncol Hematol 2020; 156:103119. [PMID: 33053439 DOI: 10.1016/j.critrevonc.2020.103119] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide. In the past decade EGFR, ALK and ROS1 TKIs lead to an unprecedented survival improvement of oncogene-addicted NSCLC patients, with better toxicity profile compared to chemotherapy. In recent years the implementation of high-throughput sequencing platforms led to the identification of uncommon molecular alterations in oncogenic drivers, such as BRAF, MET, RET, HER2 and NTRK. Moreover, newly developed drugs have been found to be active against hard to target drivers, such as KRAS. Specific TKIs targeting these genomic alterations are currently in clinical development and showed impressive activity and survival improvement, leading to FDA-accelerated approval for some of them. However, virtually all patients develop resistance to TKIs by on-target or off-target mechanisms. Here we review the clinicopathological features, the emerging targeted therapies and mechanisms of resistance and strategies to overcome them of KRAS, BRAF, MET, RET, HER2 and NTRK-addicted advanced NSCLCs.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Elisa Andrini
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Monia Sisi
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessandro Rizzo
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Claudia Parisi
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessandro Di Federico
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Francesco Gelsomino
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy.
| | - Andrea Ardizzoni
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy; Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| |
Collapse
|
807
|
Upadhya A, Yadav KS, Misra A. Targeted drug therapy in non-small cell lung cancer: Clinical significance and possible solutions-Part I. Expert Opin Drug Deliv 2020; 18:73-102. [PMID: 32954834 DOI: 10.1080/17425247.2021.1825377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) comprises of 84% of all lung cancer cases. The treatment options for NSCLC at advanced stages are chemotherapy and radiotherapy. Chemotherapy involves conventional nonspecific chemotherapeutics, and targeted-protein/receptor-specific small molecule inhibitors. Biologically targeted therapies such as an antibody-based immunotherapy have been approved in combination with conventional therapeutics. Approved targeted chemotherapy is directed against the kinase domains of mutated cellular receptors such as epidermal growth factor receptor (EGFR), anaplastic lymphoma kinases (ALK), neurotrophic receptor kinases (NTRK) and against downstream signaling molecules such as BRAF (v-raf murine sarcoma viral oncogene homolog B1). Approved biologically targeted therapy involves the use of anti-angiogenesis antibodies and antibodies against immune checkpoints. AREAS COVERED The rationale for the employment of targeted therapeutics and the resistance that may develop to therapy are discussed. Novel targeted therapeutics in clinical trials are also included. EXPERT OPINION Molecular and histological profiling of a given tumor specimen to determine the aberrant onco-driver is a must before deciding a targeted therapeutic regimen for the patient. Periodic monitoring of the patients response to a given therapeutic regimen is also mandatory so that any semblance of resistance to therapy can be deciphered and the regimen may be accordingly altered.
Collapse
Affiliation(s)
- Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| | - Ambikanandan Misra
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| |
Collapse
|
808
|
Nozaki Y, Yamamoto H, Iwasaki T, Sato M, Jiromaru R, Hongo T, Yasumatsu R, Oda Y. Clinicopathological features and immunohistochemical utility of NTRK-, ALK-, and ROS1-rearranged papillary thyroid carcinomas and anaplastic thyroid carcinomas. Hum Pathol 2020; 106:82-92. [PMID: 32980422 DOI: 10.1016/j.humpath.2020.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 02/08/2023]
Abstract
NTRK1/3, ALK, and ROS1 translocations have been reported in a minor subset of papillary thyroid carcinomas (PTCs). We aimed to elucidate the prevalence and clinicopathological characteristics of these gene rearrangements and the utility of immunohistochemistry (IHC) in PTC and anaplastic thyroid carcinoma (ATC). We screened nonradiation-exposed cases of 307 PTCs and 16 ATCs by IHC for pan-Trk, ALK, and ROS1, followed by fluorescence in situ hybridization (FISH). In the PTC group, IHC for pan-Trk, ALK, and ROS1 was positive in 18 cases (5.9%), 1 case (0.3%), and 12 cases (3.9%), respectively. Among the pan-Trk IHC-positive cases (n = 18), 2 cases (11.1%; 0.7% of all PTCs) had NTRK1 or NTRK3 gene rearrangement with conventional PTC histology. The ALK IHC-positive case (n = 1) was the follicular variant of PTC with consistent ALK gene rearrangement. ROS1 gene rearrangement was not detectable in the ROS1 IHC-positive PTCs (0/12) by FISH. Most (approximately 70%) of the pan-Trk or ROS1 IHC-positive/FISH-negative cases had BRAF gene mutation with conventional PTC morphology. In the ATC group, neither ALK nor ROS1 IHC was positive, whereas pan-Trk IHC was positive in 1 case (6.3%) in which NTRK1 gene rearrangement was confirmed by FISH. These results suggest that NTRK, ALK, and ROS1 rearrangements are rare molecular events in nonradiation-exposed Japanese patients with PTC and ATC. Although IHC is not an entirely specific surrogate for these abnormalities and does not serve as a stand-alone companion diagnosis, the combined use of IHC and molecular testing may be helpful for determining promising therapeutic strategies with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Yui Nozaki
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takeshi Iwasaki
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Masanobu Sato
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan; Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Rina Jiromaru
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan; Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takahiro Hongo
- Department of Anatomic Pathology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Ryuji Yasumatsu
- Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Otorhinolaryngology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan.
| |
Collapse
|
809
|
Walker A. Neurotrophic tyrosine kinase inhibitors: A review of implications for patients, clinicians and healthcare services. J Oncol Pharm Pract 2020; 26:2015-2019. [PMID: 32957860 DOI: 10.1177/1078155220959428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neurotrophic tyrosine receptor kinase (NTRK) inhibitors represent the latest advancement as a treatment option in targeted therapies for malignant disease. NTRK gene fusions involving NTRK1, 2 or 3 are implicated as genetics drivers for a number of tumour types which arise within adult and paedatric patients. NTRK inhibitors (Larotrectinib and Entrectinib) are effective agents which have demonstrated clinical benefit in the treatment of NTRK fusion positive solid tumours. Larotrectinib represents the first targeted agent to receive approval from international authorisation and commissioning bodies for the treatment of a specific genetic expression indiscriminate of the site from which the tumour has arisen. As such NTRK inhibitors could pave the way for international healthcare bodies to adopt a similar approach for future targeted therapies thereby altering the manner in which healthcare providers and patients are able to access and utilise innovative, targeted treatment options in future. The potential implications of this new approach are likely to impact upon several aspects of the traditional authorisation and commissioning pathways with potential changes to the design of clinical trials, the review and approval process by regulatory bodies and immunohistopathology services.
Collapse
Affiliation(s)
- Andrew Walker
- Pharmacy Department, 4014Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, UK
| |
Collapse
|
810
|
中国临床肿瘤学会非小细胞肺癌专家委员会. [Chinese Expert Consensus on Next Generation Sequencing Diagnosis
for Non-small Cell Lung Cancer (2020 Edition)]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:741-761. [PMID: 32957170 PMCID: PMC7519957 DOI: 10.3779/j.issn.1009-3419.2020.101.45] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
811
|
Sohal DPS, Kennedy EB, Cinar P, Conroy T, Copur MS, Crane CH, Garrido-Laguna I, Lau MW, Johnson T, Krishnamurthi S, Moravek C, O'Reilly EM, Philip PA, Pant S, Shah MA, Sahai V, Uronis HE, Zaidi N, Laheru D. Metastatic Pancreatic Cancer: ASCO Guideline Update. J Clin Oncol 2020; 38:3217-3230. [PMID: 32755482 DOI: 10.1200/jco.20.01364] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The aim of this work was to provide an update to the ASCO guideline on metastatic pancreatic cancer pertaining to recommendations for therapy options after first-line treatment. METHODS ASCO convened an Expert Panel and conducted a systematic review to update guideline recommendations for second-line therapy for metastatic pancreatic cancer. RESULTS One randomized controlled trial of olaparib versus placebo, one report on phase I and II studies of larotrectinib, and one report on phase I and II studies of entrectinib met the inclusion criteria and inform the guideline update. RECOMMENDATIONS New or updated recommendations for germline and somatic testing for microsatellite instability high/mismatch repair deficiency, BRCA mutations, and TRK alterations are provided for all treatment-eligible patients to select patients for recommended therapies, including pembrolizumab, olaparib, larotrectinib, or entrectinib, or potential clinical trials. The Expert Panel continues to endorse the remaining recommendations for second-line chemotherapy, as well as other recommendations related to treatment, follow-up, and palliative care from the 2018 version of this guideline. Additional information is available at www.asco.org/gastrointestinal-cancer-guidelines.
Collapse
Affiliation(s)
| | | | - Pelin Cinar
- University of California, San Francisco, San Francisco, CA
| | - Thierry Conroy
- Université de Lorraine and Institut de Cancérologie de Lorraine, Lorraine, France
| | | | | | | | | | | | | | | | | | | | | | - Manish A Shah
- New York Presbyterian/Weill Cornell Medical Center, New York, NY
| | | | | | | | - Daniel Laheru
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
812
|
The Receptor Tyrosine Kinase TrkA Is Increased and Targetable in HER2-Positive Breast Cancer. Biomolecules 2020; 10:biom10091329. [PMID: 32957504 PMCID: PMC7564775 DOI: 10.3390/biom10091329] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/21/2022] Open
Abstract
The tyrosine kinase receptor A (NTRK1/TrkA) is increasingly regarded as a therapeutic target in oncology. In breast cancer, TrkA contributes to metastasis but the clinicopathological significance remains unclear. In this study, TrkA expression was assessed via immunohistochemistry of 158 invasive ductal carcinomas (IDC), 158 invasive lobular carcinomas (ILC) and 50 ductal carcinomas in situ (DCIS). TrkA was expressed in cancer epithelial and myoepithelial cells, with higher levels of TrkA positively associated with IDC (39% of cases) (p < 0.0001). Interestingly, TrkA was significantly increased in tumours expressing the human epidermal growth factor receptor-2 (HER2), with expression in 49% of HER2-positive compared to 25% of HER2-negative tumours (p = 0.0027). A panel of breast cancer cells were used to confirm TrkA protein expression, demonstrating higher levels of TrkA (total and phosphorylated) in HER2-positive cell lines. Functional investigations using four different HER2-positive breast cancer cell lines indicated that the Trk tyrosine kinase inhibitor GNF-5837 reduced cell viability, through decreased phospho-TrkA (Tyr490) and downstream AKT (Ser473) activation, but did not display synergy with Herceptin. Overall, these data highlight a relationship between the tyrosine kinase receptors TrkA and HER2 and suggest the potential of TrkA as a novel or adjunct target for HER2-positive breast tumours.
Collapse
|
813
|
Kachko VA, Platonova NM, Vanushko VE, Shifman BM. [The role of molecular testing in thyroid tumors]. ACTA ACUST UNITED AC 2020; 66:33-46. [PMID: 33351337 DOI: 10.14341/probl12491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/14/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022]
Abstract
¹I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; ²Endocrinology Research Centre, Moscow, Russia Thyroid cancer is the most common endocrine gland cancer. In the last few decades, the molecular diagnostics for thyroid tumors have been widely researched. It is one of the few cancers whose incidence has increased in recent years from microcarcinomas to common, large forms, in all age groups, from children to the elder people. Most researches focus on the genetic basis, since our current knowledge of the genetic background of various forms of thyroid cancer is far from being complete. Molecular and genetic research has several main directions: firstly, differential diagnosis of thyroid tumors, secondly, the prognostic value of detected mutations in thyroid cancer, and thirdly, targeted therapy for aggressive or radioactive iodine-resistant forms of thyroid cancer. In this review, we wanted to update our understanding and describe the prevailing advances in molecular genetics of thyroid cancer, focusing on the main genes associated with the pathology and their potential application in clinical practice.
Collapse
Affiliation(s)
- Vera A Kachko
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | |
Collapse
|
814
|
Laskin J, Liu SV, Tolba K, Heining C, Schlenk RF, Cheema P, Cadranel J, Jones MR, Drilon A, Cseh A, Gyorffy S, Solca F, Duruisseaux M. NRG1 fusion-driven tumors: biology, detection, and the therapeutic role of afatinib and other ErbB-targeting agents. Ann Oncol 2020; 31:1693-1703. [PMID: 32916265 DOI: 10.1016/j.annonc.2020.08.2335] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/03/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022] Open
Abstract
Oncogenic gene fusions are hybrid genes that result from structural DNA rearrangements, leading to deregulated activity. Fusions involving the neuregulin-1 gene (NRG1) result in ErbB-mediated pathway activation and therefore present a rational candidate for targeted treatment. The most frequently reported NRG1 fusion is CD74-NRG1, which most commonly occurs in patients with invasive mucinous adenocarcinomas (IMAs) of the lung, although several other NRG1 fusion partners have been identified in patients with lung cancer, including ATP1B1, SDC4, and RBPMS. NRG1 fusions are also present in patients with other solid tumors, such as pancreatic ductal adenocarcinoma. In general, NRG1 fusions are rare across different types of cancer, with a reported incidence of <1%, with the notable exception of IMA, which represents ≈2%-10% of lung adenocarcinomas and has a reported incidence of ≈10%-30% for NRG1 fusions. A substantial proportion (≈20%) of NRG1 fusion-positive non-small-cell lung cancer cases are nonmucinous adenocarcinomas. ErbB-targeted treatments, such as afatinib, a pan-ErbB tyrosine kinase inhibitor, are potential therapeutic strategies to address unmet treatment needs in patients harboring NRG1 fusions.
Collapse
Affiliation(s)
- J Laskin
- Division of Medical Oncology, Department of Medicine, University of British Columbia, BC Cancer, Vancouver, BC, Canada.
| | - S V Liu
- Georgetown University Medical Center, Washington, USA
| | - K Tolba
- Oregon Health and Science University, Portland, OR, USA
| | - C Heining
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden and German Cancer Research Center (DKFZ), Dresden, Germany; Center for Personalized Oncology, NCT Dresden and University Hospital Carl Gustav Carus Dresden at Technical University Dresden, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - R F Schlenk
- National Center of Tumor Diseases Heidelberg, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - P Cheema
- William Osler Health System, University of Toronto, Toronto, ON, Canada
| | - J Cadranel
- Assistance Publique Hôpitaux de Paris, Hôpital Tenon and Sorbonne Université, Paris, France
| | - M R Jones
- QIAGEN Digital Insights, QIAGEN Inc., Redwood City, CA, USA
| | - A Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Cseh
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - S Gyorffy
- AstraZeneca Canada Ltd, Mississauga, ON, Canada
| | - F Solca
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - M Duruisseaux
- Hospices Civils de Lyon Cancer Institute, Anticancer Antibodies Lab Cancer Research Center of Lyon INSERM 1052 CNRS 528, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
815
|
Mansuet-Lupo A, Garinet S, Damotte D, Alifano M, Blons H, Wislez M, Leroy K. Les réarrangements moléculaires : cibles thérapeutiques en cancérologie thoracique. Bull Cancer 2020; 107:896-903. [DOI: 10.1016/j.bulcan.2020.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/17/2020] [Indexed: 11/16/2022]
|
816
|
The first case of gastric carcinoma with NTRK rearrangement: identification of a novel ATP1B-NTRK1 fusion. Gastric Cancer 2020; 23:944-947. [PMID: 32189226 DOI: 10.1007/s10120-020-01061-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023]
Abstract
NTRK gene rearrangements occur in a wide spectrum of tumors and are actionable events predictive of response to TRK inhibitor. We report the first case of gastric carcinoma harboring a NTRK fusion in a 79-year-old man. The tumor was composed predominantly of poorly cohesive carcinoma with focal tubular differentiation. Solid sheet-like or nested pattern of large oxyphilic cells was also noted in 10% of tumor. Pan-Trk immunohistochemistry demonstrated Trk expression with a diffuse cytoplasmic and dot-like staining only in the solid component. Extensive lymphatic invasion and multiple nodal metastases were noted and were predominated by Trk-positive component. A novel ATP1B1-NTRK1 fusion was detected by RNA-seq using fresh frozen sample. The patient died of the disease, despite surgery and chemotherapy. Although extremely rare, NTRK rearrangement does occur in gastric carcinoma and might be associated with aggressive phenotype as well as histologic features like solid growth with extensive lymphatic invasion.
Collapse
|
817
|
Liu D, Flory J, Lin A, Offin M, Falcon CJ, Murciano-Goroff YR, Rosen E, Guo R, Basu E, Li BT, Harding JJ, Iyer G, Jhaveri K, Gounder MM, Shukla NN, Roberts SS, Glade-Bender J, Kaplanis L, Schram A, Hyman DM, Drilon A. Characterization of on-target adverse events caused by TRK inhibitor therapy. Ann Oncol 2020; 31:1207-1215. [PMID: 32422171 PMCID: PMC8341080 DOI: 10.1016/j.annonc.2020.05.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The tropomyosin receptor kinase (TRK) pathway controls appetite, balance, and pain sensitivity. While these functions are reflected in the on-target adverse events (AEs) observed with TRK inhibition, these AEs remain under-recognized, and pain upon drug withdrawal has not previously been reported. As TRK inhibitors are approved by multiple regulatory agencies for TRK or ROS1 fusion-positive cancers, characterizing these AEs and corresponding management strategies is crucial. PATIENTS AND METHODS Patients with advanced or unresectable solid tumors treated with a TRK inhibitor were retrospectively identified in a search of clinical databases. Among these patients, the frequency, severity, duration, and management outcomes of AEs including weight gain, dizziness or ataxia, and withdrawal pain were characterized. RESULTS Ninety-six patients with 15 unique cancer histologies treated with a TRK inhibitor were identified. Weight gain was observed in 53% [95% confidence interval (CI), 43%-62%] of patients and increased with time on TRK inhibition. Pharmacologic intervention, most commonly with glucagon-like peptide 1 analogs or metformin, appeared to result in stabilization or loss of weight. Dizziness, with or without ataxia, was observed in 41% (95% CI, 31%-51%) of patients with a median time to onset of 2 weeks (range, 3 days to 16 months). TRK inhibitor dose reduction was the most effective intervention for dizziness. Pain upon temporary or permanent TRK inhibitor discontinuation was observed in 35% (95% CI, 24%-46%) of patients; this was more common with longer TRK inhibitor use. TRK inhibitor reinitiation was the most effective intervention for withdrawal pain. CONCLUSIONS TRK inhibition-related AEs including weight gain, dizziness, and withdrawal pain occur in a substantial proportion of patients receiving TRK inhibitors. This safety profile is unique relative to other anticancer therapies and warrants careful monitoring. These on-target toxicities are manageable with pharmacologic intervention and dose modification.
Collapse
Affiliation(s)
- D Liu
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J Flory
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - A Lin
- Department of Medicine, Weill Cornell Medical College, New York, USA; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - M Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - C J Falcon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Y R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - R Guo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Basu
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - B T Li
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - J J Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - G Iyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - K Jhaveri
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - M M Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - N N Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - S S Roberts
- Department of Medicine, Weill Cornell Medical College, New York, USA; Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J Glade-Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - L Kaplanis
- Department of Nursing, Memorial Sloan Kettering Cancer Center, New York, USA
| | - A Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - D M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - A Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA.
| |
Collapse
|
818
|
Pinheiro KV, Thomaz A, Souza BK, Metcalfe VA, Freire NH, Brunetto AT, de Farias CB, Jaeger M, Bambini V, Smith CGS, Shaw L, Roesler R. Expression and pharmacological inhibition of TrkB and EGFR in glioblastoma. Mol Biol Rep 2020; 47:6817-6828. [PMID: 32862352 DOI: 10.1007/s11033-020-05739-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
A member of the Trk family of neurotrophin receptors, tropomyosin receptor kinase B (TrkB, encoded by the NTRK2 gene) is an increasingly important target in various cancer types, including glioblastoma (GBM). EGFR is among the most frequently altered oncogenes in GBM, and EGFR inhibition has been tested as an experimental therapy. Functional interactions between EGFR and TrkB have been demonstrated. In the present study, we investigated the role of TrkB and EGFR, and their interactions, in GBM. Analyses of NTRK2 and EGFR gene expression from The Cancer Genome Atlas (TCGA) datasets showed an increase in NTRK2 expression in the proneural subtype of GBM, and a strong correlation between NTRK2 and EGFR expression in glioma CpG island methylator phenotype (G-CIMP+) samples. We showed that when TrkB and EGFR inhibitors were combined, the inhibitory effect on A172 human GBM cells was more pronounced than when either inhibitor was given alone. When U87MG GBM cells were xenografted into the flank of nude mice, tumor growth was delayed by treatment with TrkB and EGFR inhibitors, given alone or combined, only at specific time points. Intracranial GBM growth in mice was not significantly affected by drug treatments. Our findings indicate that correlations between NTRK2 and EGFR expression occur in specific GBM subgroups. Also, our results using cultured cells suggest for the first time the potential of combining TrkB and EGFR inhibition for the treatment of GBM.
Collapse
Affiliation(s)
- Kelly V Pinheiro
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), Porto Alegre, RS, 90050-170, Brazil
| | - Amanda Thomaz
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), Porto Alegre, RS, 90050-170, Brazil.,Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA 4YG, UK
| | - Bárbara Kunzler Souza
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), Porto Alegre, RS, 90050-170, Brazil.,Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Victoria Anne Metcalfe
- Faculty of Clinical and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, Lancashire, PR1 2HE, UK
| | - Natália Hogetop Freire
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - André Tesainer Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Caroline Brunetto de Farias
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Mariane Jaeger
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.,Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Victorio Bambini
- Faculty of Clinical and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, Lancashire, PR1 2HE, UK
| | - Christopher G S Smith
- Faculty of Clinical and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, Lancashire, PR1 2HE, UK
| | - Lisa Shaw
- Faculty of Clinical and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, Lancashire, PR1 2HE, UK
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil. .,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, 500 (ICBS, Campus Centro/UFRGS), Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
819
|
Cohen R, Pudlarz T, Delattre JF, Colle R, André T. Molecular Targets for the Treatment of Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:E2350. [PMID: 32825275 PMCID: PMC7563268 DOI: 10.3390/cancers12092350] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Over the past years, colorectal cancer (CRC) was subtyped according to its molecular and genetic characteristics, allowing the development of therapeutic strategies, based on predictive biomarkers. Biomarkers such as microsatellite instability (MSI), RAS and BRAF mutations, HER2 amplification or NTRK fusions represent major tools for personalized therapeutic strategies. Moreover, the routine implementation of molecular predictive tests provides new perspectives and challenges for the therapeutic management of CRC patients, such as liquid biopsies and the reintroduction of anti-EGFR monoclonal antibodies. In this review, we summarize the current landscape of targeted therapies for metastatic CRC patients, with a focus on new developments for EGFR blockade and emerging biomarkers (MSI, HER2, NTRK).
Collapse
Affiliation(s)
- Romain Cohen
- Department of Medical Oncology, Hôpital Saint-Antoine, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), F-75012 Paris, France; (T.P.); (J.-F.D.); (R.C.); (T.A.)
| | | | | | | | | |
Collapse
|
820
|
Identification of Novel Fusion Genes in Bone and Soft Tissue Sarcoma and Their Implication in the Generation of a Mouse Model. Cancers (Basel) 2020; 12:cancers12092345. [PMID: 32825119 PMCID: PMC7565474 DOI: 10.3390/cancers12092345] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/28/2022] Open
Abstract
Fusion genes induced by chromosomal aberrations are common mutations causally associated with bone and soft tissue sarcomas (BSTS). These fusions are usually disease type-specific, and identification of the fusion genes greatly helps in making precise diagnoses and determining therapeutic directions. However, there are limitations in detecting unknown fusion genes or rare fusion variants when using standard fusion gene detection techniques, such as reverse transcription-polymerase chain reaction (RT-PCR) and fluorescence in situ hybridization (FISH). In the present study, we have identified 19 novel fusion genes using target RNA sequencing (RNA-seq) in 55 cases of round or spindle cell sarcomas in which no fusion genes were detected by RT-PCR. Subsequent analysis using Sanger sequencing confirmed that seven out of 19 novel fusion genes would produce functional fusion proteins. Seven fusion genes detected in this study affect signal transduction and are ideal targets of small molecule inhibitors. YWHAE-NTRK3 expression in mouse embryonic mesenchymal cells (eMCs) induced spindle cell sarcoma, and the tumor was sensitive to the TRK inhibitor LOXO-101 both in vitro and in vivo. The combination of target RNA-seq and generation of an ex vivo mouse model expressing novel fusions provides important information both for sarcoma biology and the appropriate diagnosis of BSTS.
Collapse
|
821
|
Abstract
PURPOSE OF REVIEW We herein review some of the major patterns of resistance and lessons learned from the use of earlier targeted therapies in two genotype-driven solid tumors. RECENT FINDINGS Targeted agents have rapidly expanded in the field of oncology over the past 2 decades. The breakthroughs achieved by these agents have been, however, hindered by the inevitable development of drug resistance. Intrinsic or acquired mechanisms of resistance eventually lead to treatment tolerance and tumoral plasticity with phenotypic switch and evasion of the original targeted pathway. Failures in such therapies also result from poor selectivity of the target, drug delivery, and unaffordable costs. SUMMARY Based on above findings, collaborative efforts are advancing at the molecular level to design better drugs or combinatorial strategies and to develop more sensitive assays to monitor responses and the emergence of resistance.
Collapse
|
822
|
Swan K, Dougherty KC, Myers SW. Somatic Testing and Germline Genetic Status: Implications for Cancer Treatment Decisions and Genetic Counseling. CURRENT GENETIC MEDICINE REPORTS 2020. [DOI: 10.1007/s40142-020-00192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
823
|
Abstract
Almost all uterine mesenchymal tumours have been historically classified as either smooth muscle or endometrial stromal neoplasms. Recent application of molecular techniques has identified numerous lesions with distinctive genetic abnormalities and clinicopathological characteristics. Newly discovered uterine sarcoma subtypes include high-grade endometrial stromal sarcomas with BCOR genetic abnormalities, fibrosarcoma-like uterine sarcomas with NTRK rearrangements and COL1A-PDGFRB fusions, as well as undifferentiated uterine sarcomas with SMARCA4 mutations. Novel PLAG1 and PGR fusions have been identified in subsets of myxoid and epithelioid leiomyosarcomas. Some uterine tumours resembling ovarian sex-cord tumour harbour GREB1 and ESR1 rearrangements. Histological and immunophenotypical features as well as underlying genetic abnormalities defining these lesions are discussed.
Collapse
Affiliation(s)
| | - Sarah Chiang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
824
|
Guo Y, Guo X, Wang S, Chen X, Shi J, Wang J, Wang K, Klempner SJ, Wang W, Xiao M. Genomic Alterations of NTRK, POLE, ERBB2, and Microsatellite Instability Status in Chinese Patients with Colorectal Cancer. Oncologist 2020; 25:e1671-e1680. [PMID: 32627883 PMCID: PMC7648350 DOI: 10.1634/theoncologist.2020-0356] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022] Open
Abstract
Background The increasing molecular characterization of colorectal cancers (CRCs) has spurred the need to look beyond RAS, BRAF, and microsatellite instability (MSI). Genomic alterations, including ERBB2 amplifications and mutations, POLE mutations, MSI, and NTRK1–3 fusions, have emerged as targets for matched therapies. We sought to study a clinically annotated Chinese cohort of CRC subjected to genomic profiling to explore relative target frequencies. Methods Tumor and matched whole blood were collected from 609 Chinese patients with CRC. Extracted DNA was analyzed for all classes of genomic alterations across 450 cancer‐related genes, including single‐nucleotide variations (SNVs), short and long insertions and deletions (indels), copy number variations, and gene rearrangements. Next‐generation sequencing–based computational algorithms also determined tumor mutational burden and MSI status. Results Alterations in TP53 (76%), APC (72%), and KRAS (46%) were common in Chinese patients with CRC. For the first time, the prevalence of NTRK gene fusion was observed to be around 7% in the MSI‐high CRC cohort. Across the cohort, MSI was found in 9%, ERBB2 amplification in 3%, and POLE pathogenic mutation in 1.5% of patients. Such results mostly parallel frequencies observed in Western patients. However, POLE existed at a higher frequency and was associated with large tumor T‐cell infiltration. Conclusion Comparing to the Western counterparts, POLE mutations were increased in our cohort. The prevalence of NTRK gene fusion was around 7% in the MSI‐high CRC cohort. Increased adoption of molecular profiling in Asian patients is essential for the improvement of therapeutic outcomes. Implications for Practice The increasing use of genomic profiling assays in colorectal cancer (CRC) has allowed for the identification of a higher number of patient subsets benefiting from matched therapies. With an increase in the number of therapies, assays simultaneously evaluating all candidate biomarkers are critical. The results of this study provide an early support for the feasibility and utility of genomic profiling in Chinese patients with CRC. The emergence of precision medicine has identified genomic variants, such as NTRK gene fusion, microsatellite instability (MSI), HER2 amplification, and POLE pathogenic mutation, as potential agonistic biomarkers for immune or targeted therapies. This article examines NTRK, HER2, and POLE in a cohort of Chinese patients with colorectal cancer.
Collapse
Affiliation(s)
- Yun Guo
- First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Xian‐ling Guo
- Department of Medical Oncology, 10th People's Hospital, Tongji UniversityShanghaiPeople's Republic of China
- Department of Medical Oncology, Dermatology Hospital, Tongji UniversityShanghaiPeople's Republic of China
| | - Shuang Wang
- Nanfang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Xinyu Chen
- First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | | | - Jian Wang
- OrigiMedShanghaiPeople's Republic of China
| | - Kai Wang
- OrigiMedShanghaiPeople's Republic of China
| | - Samuel J. Klempner
- Department of Medicine, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Min Xiao
- Shu Lan (Hangzhou) HospitalHangzhouPeople's Republic of China
| |
Collapse
|
825
|
Dupuis M, Shen Y, Curcio C, Meis JM, Wang WL, Amini B, Rhines L, Reuther J, Roy A, Fisher KE, Conley AP, Andrew Livingston J. Successful treatment of lipofibromatosis-like neural tumor of the lumbar spine with an NTRK-fusion inhibitor. Clin Sarcoma Res 2020; 10:14. [PMID: 32782782 PMCID: PMC7412810 DOI: 10.1186/s13569-020-00136-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background Lipofibromatosis-like neural tumors (LPF-NT) are a newly identified class of rare mesenchymal neoplasms. Current standard of care therapy is surgical resection alone; there are no chemotherapies or molecular targeted therapies that have been shown to be effective in patients who are not surgical candidates due to either tumor bulk or location. Most LPF-NT harbor NTRK fusions, although the therapeutic significance of these fusions has not been previously demonstrated in this malignancy. Here, we present the first case of a patient with surgically-unresectable LPF-NT successfully treated with medical therapy, specifically the TRK fusion-protein inhibitor entrectinib. Case presentation The patient is a 21 year old man with no co-morbidities who presented for evaluation due to intermittent abdominal pain and was found to have a mass spanning from T12-L2. Biopsy revealed a mesenchymal spindle cell neoplasm and S100 positivity pointed to possible nerve sheath origin. The sample was ultimately found to have an LMNA-NTRK1 fusion, confirming the diagnosis of LP-NFT. Unfortunately, due to the bulk and location of the tumor, surgery was felt to be exceptionally morbid and the patient was treated in a clinical trial with the NTRK inhibitor entrectinib. Surprisingly, he had such a robust clinical response that he was ultimately deemed a surgical candidate and he was successfully taken to surgery. Post-operative pathology revealed > 95% necrosis, demonstrating exceptional sensitivity to the targeted therapy. The patient remains NED and on entrectinib 12 months post-operatively. Conclusions The exceptional treatment response of this patient suggests that NTRK fusions are true drivers of the disease. Thus, all patients should be evaluated for NTRK fusions using sensitive methodologies and treatment with TRK fusion-protein inhibitors should be considered in patients who are not candidates for oncologic resection.
Collapse
Affiliation(s)
- Megan Dupuis
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Yulei Shen
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX USA
| | - Christian Curcio
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jeanne M Meis
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Wei-Lien Wang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Behrang Amini
- Department of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Laurence Rhines
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jacquelyn Reuther
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX USA
| | - Angshumoy Roy
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX USA
| | - Kevin E Fisher
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX USA
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0450, Houston, TX 77030 USA
| | - J Andrew Livingston
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0450, Houston, TX 77030 USA.,Department of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
826
|
Nylén C, Mechera R, Maréchal-Ross I, Tsang V, Chou A, Gill AJ, Clifton-Bligh RJ, Robinson BG, Sywak MS, Sidhu SB, Glover AR. Molecular Markers Guiding Thyroid Cancer Management. Cancers (Basel) 2020; 12:cancers12082164. [PMID: 32759760 PMCID: PMC7466065 DOI: 10.3390/cancers12082164] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
The incidence of thyroid cancer is rapidly increasing, mostly due to the overdiagnosis and overtreatment of differentiated thyroid cancer (TC). The increasing use of potent preclinical models, high throughput molecular technologies, and gene expression microarrays have provided a deeper understanding of molecular characteristics in cancer. Hence, molecular markers have become a potent tool also in TC management to distinguish benign from malignant lesions, predict aggressive biology, prognosis, recurrence, as well as for identification of novel therapeutic targets. In differentiated TC, molecular markers are mainly used as an adjunct to guide management of indeterminate nodules on fine needle aspiration biopsies. In contrast, in advanced thyroid cancer, molecular markers enable targeted treatments of affected signalling pathways. Identification of the driver mutation of targetable kinases in advanced TC can select treatment with mutation targeted tyrosine kinase inhibitors (TKI) to slow growth and reverse adverse effects of the mutations, when traditional treatments fail. This review will outline the molecular landscape and discuss the impact of molecular markers on diagnosis, surveillance and treatment of differentiated, poorly differentiated and anaplastic follicular TC.
Collapse
Affiliation(s)
- Carolina Nylén
- Endocrine Surgical Unit, Royal North Shore Hospital, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; (C.N.); (R.M.); (M.S.S.); (S.B.S.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna L1:00, 171 76 Stockholm, Sweden
| | - Robert Mechera
- Endocrine Surgical Unit, Royal North Shore Hospital, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; (C.N.); (R.M.); (M.S.S.); (S.B.S.)
- Department of Visceral Surgery, Clarunis University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Isabella Maréchal-Ross
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
| | - Venessa Tsang
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- Department of Endocrinology, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW 2065, Australia
| | - Angela Chou
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW 2065, Australia
| | - Anthony J. Gill
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW 2065, Australia
| | - Roderick J. Clifton-Bligh
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- Department of Endocrinology, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW 2065, Australia
- Cancer Genetics Unit, Kolling Institute, Sydney, NSW 2010, Australia
| | - Bruce G. Robinson
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- Department of Endocrinology, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW 2065, Australia
- Cancer Genetics Unit, Kolling Institute, Sydney, NSW 2010, Australia
| | - Mark S. Sywak
- Endocrine Surgical Unit, Royal North Shore Hospital, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; (C.N.); (R.M.); (M.S.S.); (S.B.S.)
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
| | - Stan B. Sidhu
- Endocrine Surgical Unit, Royal North Shore Hospital, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; (C.N.); (R.M.); (M.S.S.); (S.B.S.)
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- Cancer Genetics Unit, Kolling Institute, Sydney, NSW 2010, Australia
| | - Anthony R. Glover
- Endocrine Surgical Unit, Royal North Shore Hospital, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; (C.N.); (R.M.); (M.S.S.); (S.B.S.)
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (I.M.-R.); (V.T.); (A.C.); (A.J.G.); (R.J.C.-B.); (B.G.R.)
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Faculty of Medicine, St. Vincent’s Clinical School, University of New South Wales Sydney, Sydney, NSW 2010, Australia
- Correspondence: ; Tel.: +61-2-9463-1477
| |
Collapse
|
827
|
Colomer R, Mondejar R, Romero-Laorden N, Alfranca A, Sanchez-Madrid F, Quintela-Fandino M. When should we order a next generation sequencing test in a patient with cancer? EClinicalMedicine 2020; 25:100487. [PMID: 32775973 PMCID: PMC7397394 DOI: 10.1016/j.eclinm.2020.100487] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/12/2020] [Accepted: 07/16/2020] [Indexed: 12/31/2022] Open
Abstract
Technical advances in genome sequencing and the implementation of next-generation sequencing (NGS) in clinical oncology have paved the way for individualizing cancer patient therapy based on molecular profiles. When and how to use NGS testing in the clinic is at present an unsolved issue, although new research results provide evidence favoring this approach in some types of advanced cancer. Clinical research is evolving rapidly, from basket and umbrella trials to adaptative design precision oncology clinical studies, and genomic and molecular data often displace the classical clinical validation procedures of biomarkers. In this context, physicians must be aware of the clinical evidence behind these new biomarkers and NGS tests available, in order to use them in the right moment, and with a critical point of view. This review will present the status of currently available targeted drugs that can be effective based on actionable molecular alterations, and the NGS tests that are currently available, offering a practical guide for the application of Clinical Precision Oncology in the real world routine practice.
Collapse
Affiliation(s)
- Ramon Colomer
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Endowed Chair of Personalised Precision Medicine, Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Rebeca Mondejar
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Endowed Chair of Personalised Precision Medicine, Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche, Spain
| | - Nuria Romero-Laorden
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | | | - Francisco Sanchez-Madrid
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Endowed Chair of Personalised Precision Medicine, Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche, Spain
- Immunology Division, Hospital Universitario La Princesa, Madrid, Spain
| | - Miguel Quintela-Fandino
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Medical Oncology Division, Hospital Universitario Quirón, Pozuelo de Alarcón – Madrid, Spain
| |
Collapse
|
828
|
Roviello G, D'Angelo A, Sciortino M, Mini E, Nobili S, De Logu F, Massi D. TRK fusion positive cancers: From first clinical data of a TRK inhibitor to future directions. Crit Rev Oncol Hematol 2020; 152:103011. [PMID: 32521311 DOI: 10.1016/j.critrevonc.2020.103011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022] Open
Abstract
Genetic alterations of neurotrophic tropomyosin or tyrosine receptor kinase (NTRK) 1/2/3 genes generate TRK fusion proteins have been reported in a variety of adult and child cancers from diverse cell/tissue lineages. Larotrectinib, a tumour-agnostic TRK inhibitor, has shown remarkable efficacy in a novel "basket" study which has enrolled patients from infants to elderly with different TRK fusion-positive cancers. In this review, we focus on the challenges and expectations on the development of "tumour-agnostic" targeted therapies in rare malignancies.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy.
| | - Alberto D'Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, United Kingdom
| | | | - Enrico Mini
- Department of Health Sciences, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniela Massi
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence and Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| |
Collapse
|
829
|
Lee J, Park S, Jung HA, Sun JM, Lee SH, Ahn JS, Park K, Ahn MJ. Evaluating entrectinib as a treatment option for non-small cell lung cancer. Expert Opin Pharmacother 2020; 21:1935-1942. [PMID: 32736487 DOI: 10.1080/14656566.2020.1798932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Entrectinib, an oral pan-TRK, ALK, and ROS1 inhibitor is approved as a first-line treatment for NTRK-rearranged solid tumors and ROS1-rearranged non-small cell lung cancer (NSCLC). It has demonstrated clinical efficacy for patients harboring the relevant gene rearrangement in both systemic and intracranial disease, regardless of the tumor type. AREAS COVERED In this review, the authors analyzed data from preclinical and clinical studies, the characteristics of entrectinib compared to those of other relevant inhibitors (currently available and/or under investigation), and the emerging resistance mechanisms. The authors then provide the readers with their future perspectives. EXPERT OPINION Entrectinib has been well studied across many tumor types, including NSCLC with ALK, ROS1, and NTRK rearrangements. The drug has demonstrated favorable properties with oral administration, prolonged response duration, high intracranial efficacy, and a favorable toxicity profile. However, with acquisition of resistance and the development of newer generation TKIs, the optimal place for entrectinib in the landscape of targeted therapies for NSCLC warrants further validation.
Collapse
Affiliation(s)
- Jiyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| |
Collapse
|
830
|
Yang SR, Schultheis AM, Yu H, Mandelker D, Ladanyi M, Büttner R. Precision medicine in non-small cell lung cancer: Current applications and future directions. Semin Cancer Biol 2020; 84:184-198. [PMID: 32730814 DOI: 10.1016/j.semcancer.2020.07.009] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/24/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022]
Abstract
Advances in biomarkers, targeted therapies, and immuno-oncology have transformed the clinical management of patients with advanced NSCLC. For oncogene-driven tumors, there are highly effective targeted therapies against EGFR, ALK, ROS1, BRAF, TRK, RET, and MET. In addition, investigational therapies for KRAS, NRG1, and HER2 have shown promising results and may become standard-of-care in the near future. In parallel, immune-checkpoint therapy has emerged as an indispensable treatment modality, especially for patients lacking actionable oncogenic drivers. While PD-L1 expression has shown modest predictive utility, biomarkers for immune-checkpoint inhibition in NSCLC have remained elusive and represent an area of active investigation. Given the growing importance of biomarkers, optimal utilization of small tissue biopsies and alternative genotyping methods using circulating cell-free DNA have become increasingly integrated into clinical practice. In this review, we will summarize the current landscape and emerging trends in precision medicine for patients with advanced NSCLC with a special focus on predictive biomarker testing.
Collapse
Affiliation(s)
- Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | | | - Helena Yu
- Memorial Sloan Kettering Cancer Center, Department of Medicine, United States
| | - Diana Mandelker
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | - Reinhard Büttner
- University Hospital of Cologne, Department of Pathology, Germany.
| |
Collapse
|
831
|
Dong D, Shen G, Da Y, Zhou M, Yang G, Yuan M, Chen R. Successful Treatment of Patients with Refractory High-Grade Serous Ovarian Cancer with GOPC-ROS1 Fusion Using Crizotinib: A Case Report. Oncologist 2020; 25:e1720-e1724. [PMID: 32652753 DOI: 10.1634/theoncologist.2019-0609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Recently, multiple poly (ADP-ribose) polymerase (PARP) inhibitors have demonstrated excellent efficacy among patients with ovarian cancer with or without BRCA mutations. However, alternative therapeutic options are urgently required for patients who cannot benefit from conventional chemotherapy or PARP inhibitors. CASE PRESENTATION A patient with high-grade serous ovarian carcinoma presented to our clinic after developing resistance to chemotherapy. Paired tumor-normal next-generation sequencing (NGS) was performed using peripheral blood to identify potential actionable mutations. NGS revealed the patient harboring a GOPC-ROS1 fusion, which was subsequently verified using a reverse transcription polymerase chain reaction assay. No germline or somatic mutation in BRCA1/2 or mismatch repair genes was detected. Therefore, the patient received crizotinib treatment. A rapid, favorable clinical response (partial response at 1 month) was observed, with further pathological response monitored and evaluated in follow-up interrogation. CONCLUSION This study suggested that crizotinib was an off-the-shelf, practical, and ostensibly effective treatment option for patients with ovarian cancer with ROS1 rearrangement. NGS-based genetic testing may guide to plan therapeutic paradigms, and render precision medicine promising in ovarian cancer treatment. IMPLICATIONS FOR PRACTICE Despite the previous report of ROS1 fusion in patients with ovarian cancer, it remains unknown whether patients can benefit from targeted therapeutic drugs. This study reports a GOPC-ROS1 fusion identified by next-generation sequencing in a patient with chemotherapy-resistant ovarian cancer. The patient was administered crizotinib and showed rapid, remarkable response. This study suggests that comprehensive sequencing should be offered for patients with ovarian cancer without effective therapeutic strategies, and crizotinib can be used to treat ROS1-rearranged ovarian carcinomas.
Collapse
Affiliation(s)
- Dapeng Dong
- Department of Oncology, Beijing Hui 'an Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, People's Republic of China
- Department of Oncology, Beijing Fengtai Youanmen Hospital, Beijing, People's Republic of China
| | - Ge Shen
- Department of Oncology, Beijing Hui 'an Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, People's Republic of China
- Department of Oncology, Beijing Fengtai Youanmen Hospital, Beijing, People's Republic of China
| | - Yong Da
- Department of Oncology, Beijing Hui 'an Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, People's Republic of China
- Department of Oncology, Beijing Fengtai Youanmen Hospital, Beijing, People's Republic of China
| | - Ming Zhou
- Department of Oncology, Beijing Hui 'an Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, People's Republic of China
- Department of Oncology, Beijing Fengtai Youanmen Hospital, Beijing, People's Republic of China
| | - Gang Yang
- Department of Oncology, Beijing Hui 'an Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, People's Republic of China
- Department of Oncology, Beijing Fengtai Youanmen Hospital, Beijing, People's Republic of China
| | - Mingming Yuan
- Geneplus-Beijing Ltd., Beijing, People's Republic of China
| | - Rongrong Chen
- Geneplus-Beijing Ltd., Beijing, People's Republic of China
| |
Collapse
|
832
|
Song Z, Xu C, He Y, Li F, Wang W, Zhu Y, Gao Y, Ji M, Chen M, Lai J, Cheng W, Benes CH, Chen L. Simultaneous Detection of Gene Fusions and Base Mutations in Cancer Tissue Biopsies by Sequencing Dual Nucleic Acid Templates in Unified Reaction. Clin Chem 2020; 66:178-187. [PMID: 31810998 DOI: 10.1373/clinchem.2019.308833] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/07/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Targeted next-generation sequencing is a powerful method to comprehensively identify biomarkers for cancer. Starting material is currently either DNA or RNA for different variations, but splitting to 2 assays is burdensome and sometimes unpractical, causing delay or complete lack of detection of critical events, in particular, potent and targetable fusion events. An assay that analyzes both templates in a streamlined process is eagerly needed. METHODS We developed a single-tube, dual-template assay and an integrated bioinformatics pipeline for relevant variant calling. RNA was used for fusion detection, whereas DNA was used for single-nucleotide variations (SNVs) and insertion and deletions (indels). The reaction chemistry featured barcoded adaptor ligation, multiplexed linear amplification, and multiplexed PCR for noise reduction and novel fusion detection. An auxiliary quality control assay was also developed. RESULTS In a 1000-sample lung tumor cohort, we identified all major SNV/indel hotspots and fusions, as well as MET exon 14 skipping and several novel or rare fusions. The occurrence frequencies were in line with previous reports and were verified by Sanger sequencing. One noteworthy fusion event was HLA-DRB1-MET that constituted the second intergenic MET fusion ever detected in lung cancer. CONCLUSIONS This method should benefit not only a majority of patients carrying core actionable targets but also those with rare variations. Future extension of this assay to RNA expression and DNA copy number profiling of target genes such as programmed death-ligand 1 may provide additional biomarkers for immune checkpoint therapies.
Collapse
Affiliation(s)
- Zhengbo Song
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, PR China
| | - Chunwei Xu
- Department of Pathology, Fujian Cancer Hospital and Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Yunwei He
- HeliTec Biotechnologies, Shenzhen, Guangdong Province, PR China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan, Guangdong Province, PR China
| | - Wenxian Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, PR China
| | - Youcai Zhu
- Department of Thoracic Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang Province, PR China
| | - Yanqiu Gao
- HeliTec Biotechnologies, Shenzhen, Guangdong Province, PR China
| | - Mingfang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan, Guangdong Province, PR China
| | - Miao Chen
- HeliTec Biotechnologies, Shenzhen, Guangdong Province, PR China
| | - Jiajia Lai
- HeliTec Biotechnologies, Shenzhen, Guangdong Province, PR China
| | - Weimin Cheng
- Cancer Research Institute of Zhongshan City, Zhongshan, Guangdong Province, PR China
| | - Cyril H Benes
- Massachusetts General Hospital Cancer Research Center and Harvard Medical School, Charlestown, MA
| | - Li Chen
- HeliTec Biotechnologies, Shenzhen, Guangdong Province, PR China
| |
Collapse
|
833
|
Martini G, Dienstmann R, Ros J, Baraibar I, Cuadra-Urteaga JL, Salva F, Ciardiello D, Mulet N, Argiles G, Tabernero J, Elez E. Molecular subtypes and the evolution of treatment management in metastatic colorectal cancer. Ther Adv Med Oncol 2020; 12:1758835920936089. [PMID: 32782486 PMCID: PMC7383645 DOI: 10.1177/1758835920936089] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease representing a therapeutic challenge, which is further complicated by the common occurrence of several molecular alterations that confer resistance to standard chemotherapy and targeted agents. Mechanisms of resistance have been identified at multiple levels in the epidermal growth factor receptor (EGFR) pathway, including mutations in KRAS, NRAS, and BRAF V600E, and in the HER2 and MET receptors. These alterations represent oncogenic drivers that may co-exist in the same tumor with other primary and acquired alterations via a clonal selection process. Other molecular alterations include DNA damage repair mechanisms and rare kinase fusions, potentially offering a rationale for new therapeutic strategies. In recent years, genomic analysis has been expanded by a more complex study of epigenomic, transcriptomic, and microenvironment features. The Consensus Molecular Subtype (CMS) classification describes four CRC subtypes with distinct biological characteristics that show prognostic and potential predictive value in the clinical setting. Here, we review the panorama of actionable targets in CRC, and the developments in more recent molecular tests, such as liquid biopsy analysis, which are increasingly offering clinicians a means of ensuring optimal tailored treatments for patients with metastatic CRC according to their evolving molecular profile and treatment history.
Collapse
Affiliation(s)
- Giulia Martini
- Università della Campania L. Vanvitelli, Naples
- Vall d’Hebron Institute of Oncology, P/ Vall D’Hebron 119-121, Barcelona, 08035, Spain
| | | | - Javier Ros
- Vall d’Hebron Hospital, Barcelona, Catalunya, Spain
| | | | | | | | - Davide Ciardiello
- Università della Campania L. Vanvitelli, Naples
- Vall d’Hebron Hospital, Barcelona, Catalunya, Spain
| | - Nuria Mulet
- Vall d’Hebron Hospital, Barcelona, Catalunya, Spain
| | | | | | - Elena Elez
- Vall D’Hebron Institute of Oncology P/Vall D’Hebron 119-121, Barcelona, 08035 Spain
| |
Collapse
|
834
|
Ward RA, Fawell S, Floc'h N, Flemington V, McKerrecher D, Smith PD. Challenges and Opportunities in Cancer Drug Resistance. Chem Rev 2020; 121:3297-3351. [PMID: 32692162 DOI: 10.1021/acs.chemrev.0c00383] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been huge progress in the discovery of targeted cancer therapies in recent years. However, even for the most successful and impactful cancer drugs which have been approved, both innate and acquired mechanisms of resistance are commonplace. These emerging mechanisms of resistance have been studied intensively, which has enabled drug discovery scientists to learn how it may be possible to overcome such resistance in subsequent generations of treatments. In some cases, novel drug candidates have been able to supersede previously approved agents; in other cases they have been used sequentially or in combinations with existing treatments. This review summarizes the current field in terms of the challenges and opportunities that cancer resistance presents to drug discovery scientists, with a focus on small molecule therapeutics. As part of this review, common themes and approaches have been identified which have been utilized to successfully target emerging mechanisms of resistance. This includes the increase in target potency and selectivity, alternative chemical scaffolds, change of mechanism of action (covalents, PROTACs), increases in blood-brain barrier permeability (BBBP), and the targeting of allosteric pockets. Finally, wider approaches are covered such as monoclonal antibodies (mAbs), bispecific antibodies, antibody drug conjugates (ADCs), and combination therapies.
Collapse
Affiliation(s)
- Richard A Ward
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Stephen Fawell
- Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Nicolas Floc'h
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | | | | | - Paul D Smith
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| |
Collapse
|
835
|
Goh XN, Seng MSF, Loh AHP, Gupta A, Chang KTE, Iyer P. Larotrectinib followed by selitrectinib in a novel DCTN1-NTRK1 fusion undifferentiated pleomorphic sarcoma. J Oncol Pharm Pract 2020; 27:485-489. [PMID: 32693686 DOI: 10.1177/1078155220938849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Neurotrophic receptor tyrosine kinase fusions cause overexpression or activation of kinase and are believed to confer oncogenic potential in some non-rhabdomyosarcoma soft tissue sarcomas. TRK inhibitors have recently been shown to induce responses in these tumours though current experience with these agents is still limited. CASE REPORT We report a case of an adolescent with treatment-refractory non-rhabdomyosarcoma soft tissue sarcomas, carrying a novel DCTN1-NTRK1 gene fusion whose progressive disease was treated with multi-kinase and TRK inhibitors.Management and outcome: Our patient was started on pan-TRK inhibitor larotrectinib, as his disease progressed after chemotherapy, radiation therapy and surgery, based on next-generation sequencing test showing DCTN1-NTRK1 gene fusion. He responded quickly to larotrectinib with the improvement of symptoms and reduction of masses. However, this response was short-lived due to the development of acquired solvent front resistance mutation. This patient did not respond to next-generation TRK inhibitor selitrectinib and eventually succumbed to his disease. DISCUSSION The initial rapid and drastic response of our patient to larotrectinib was not sustained due to the development of acquired resistance. This case emphasizes the need for upfront and periodic next-generation sequencing testing to guide treatment of patients with refractory non-rhabdomyosarcoma soft tissue sarcomas.
Collapse
Affiliation(s)
- Xue Na Goh
- Department of Pharmacy, KK Women's and Children's Hospital, Singapore, Singapore
| | - Michaela Su-Fern Seng
- Department of Paediatric Subspecialties, Haematology-Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Amos Hong Pheng Loh
- Duke-NUS Medical School, Singapore, Singapore
- Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore, Singapore
| | - Achint Gupta
- Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital, Singapore, Singapore
| | - Kenneth Tou En Chang
- Duke-NUS Medical School, Singapore, Singapore
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Prasad Iyer
- Department of Paediatric Subspecialties, Haematology-Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
836
|
Cationic Pillar[6]arene Induces Cell Apoptosis by Inhibiting Protein Tyrosine Phosphorylation Via Host-Guest Recognition. Int J Mol Sci 2020; 21:ijms21144979. [PMID: 32679647 PMCID: PMC7404071 DOI: 10.3390/ijms21144979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022] Open
Abstract
We reported for the first time that cationic pillar[6]arene (cPA6) could tightly bind to peptide polymer (MW~20–50 kDa), an artificial substrate for tyrosine (Tyr) phosphorylation, and efficiently inhibit Tyr protein phosphorylation through host–guest recognition. We synthesized a nanocomposite of black phosphorus nanosheets loaded with cPA6 (BPNS@cPA6) to explore the effect of cPA6 on cells. BPNS@cPA6 was able to enter HepG2 cells, induced apoptosis, and inhibited cell proliferation by reducing the level of Tyr phosphorylation. Furthermore, BPNS@cPA6 showed a stronger ability of inhibiting cell proliferation in tumor cells than in normal cells. Our results revealed the supramolecular modulation of enzymatic Tyr phosphorylation by the host–guest recognition of cPA6.
Collapse
|
837
|
Wei D, Qi C, Wu Y, Zhang X, Ren G. AKAP13-NTRK3: A novel NTRK3 oncogenic fusion variant in a patient with melanoma. Oral Oncol 2020; 111:104891. [PMID: 32654869 DOI: 10.1016/j.oraloncology.2020.104891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Dongliang Wei
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China
| | - Chuang Qi
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, China
| | - Yuan Wu
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, China
| | - Xing Zhang
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, China
| | - Guoxin Ren
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China.
| |
Collapse
|
838
|
Torre M, Vasudevaraja V, Serrano J, DeLorenzo M, Malinowski S, Blandin AF, Pages M, Ligon AH, Dong F, Meredith DM, Nasrallah MP, Horbinski C, Dahiya S, Ligon KL, Santi M, Ramkissoon SH, Filbin MG, Snuderl M, Alexandrescu S. Molecular and clinicopathologic features of gliomas harboring NTRK fusions. Acta Neuropathol Commun 2020; 8:107. [PMID: 32665022 PMCID: PMC7362646 DOI: 10.1186/s40478-020-00980-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023] Open
Abstract
Fusions involving neurotrophic tyrosine receptor kinase (NTRK) genes are detected in ≤2% of gliomas and can promote gliomagenesis. The remarkable therapeutic efficacy of TRK inhibitors, which are among the first Food and Drug Administration-approved targeted therapies for NTRK-fused gliomas, has generated significant clinical interest in characterizing these tumors. In this multi-institutional retrospective study of 42 gliomas with NTRK fusions, next generation DNA sequencing (n = 41), next generation RNA sequencing (n = 1), RNA-sequencing fusion panel (n = 16), methylation profile analysis (n = 18), and histologic evaluation (n = 42) were performed. All infantile NTRK-fused gliomas (n = 7) had high-grade histology and, with one exception, no other significant genetic alterations. Pediatric NTRK-fused gliomas (n = 13) typically involved NTRK2, ranged from low- to high-histologic grade, and demonstrated histologic overlap with desmoplastic infantile ganglioglioma, pilocytic astrocytoma, ganglioglioma, and glioblastoma, among other entities, but they rarely matched with high confidence to known methylation class families or with each other; alterations involving ATRX, PTEN, and CDKN2A/2B were present in a subset of cases. Adult NTRK-fused gliomas (n = 22) typically involved NTRK1 and had predominantly high-grade histology; genetic alterations involving IDH1, ATRX, TP53, PTEN, TERT promoter, RB1, CDKN2A/2B, NF1, and polysomy 7 were common. Unsupervised principal component analysis of methylation profiles demonstrated no obvious grouping by histologic grade, NTRK gene involved, or age group. KEGG pathway analysis detected methylation differences in genes involved in PI3K/AKT, MAPK, and other pathways. In summary, the study highlights the clinical, histologic, and molecular heterogeneity of NTRK-fused gliomas, particularly when stratified by age group.
Collapse
Affiliation(s)
- Matthew Torre
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, 300 Longwood Ave, Bader Building, Boston, MA 02115 USA
| | - Varshini Vasudevaraja
- Department of Pathology, NYU Langone Health, 550 First Avenue, New York, NY 10016 USA
| | - Jonathan Serrano
- Department of Pathology, NYU Langone Health, 550 First Avenue, New York, NY 10016 USA
| | - Michael DeLorenzo
- Department of Pathology, NYU Langone Health, 550 First Avenue, New York, NY 10016 USA
| | - Seth Malinowski
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02115 USA
| | - Anne-Florence Blandin
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02115 USA
| | - Melanie Pages
- Department of Neuropathology, GHU Paris Sainte-Anne Hospital, 1 Rue Cabanis, 75014 Paris, France
| | - Azra H. Ligon
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
- Center for Advanced Molecular Diagnostics, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Fei Dong
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - David M. Meredith
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - MacLean P. Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street 34th St, Philadelphia, PA 19104 USA
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, IL USA
- Department of Pathology, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611 USA
| | - Sonika Dahiya
- Division of Neuropathology, Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8118, St. Louis, MO 63110 USA
| | - Keith L. Ligon
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, 300 Longwood Ave, Bader Building, Boston, MA 02115 USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02115 USA
| | - Mariarita Santi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street 34th St, Philadelphia, PA 19104 USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Shakti H. Ramkissoon
- Foundation Medicine, 7010 Kit Creek Road, Morrisville, NC 27560 USA
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, 27157 NC USA
| | - Mariella G. Filbin
- Department of Pediatric Oncology, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215 USA
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health, 550 First Avenue, New York, NY 10016 USA
| | - Sanda Alexandrescu
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, 300 Longwood Ave, Bader Building, Boston, MA 02115 USA
| |
Collapse
|
839
|
Forschner A, Forchhammer S, Bonzheim I. NTRK gene fusions in melanoma: detection, prevalence and potential therapeutic implications. J Dtsch Dermatol Ges 2020; 18:1387-1392. [PMID: 32656925 DOI: 10.1111/ddg.14160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023]
Abstract
Fusions involving neurotrophic tyrosine receptor kinase (NTRK) are known drivers of oncogenesis and also occur in melanoma, although very rarely. A particularly high incidence of NTRK gene fusions is reported in infantile fibrosarcoma (> 90 %) or the secretory type of breast cancer (> 90 %). Recently, larotrectinib (a tropomyosin receptor kinase [TRK] inhibitor) was approved, and we wondered whether TRK inhibitors might also be helpful for melanoma patients. We therefore screened the literature and obtained relevant results. NTRK fusions are relatively common in spitzoid melanoma, with a prevalence of 21-29 % compared to < 1 % in cutaneous or mucosal melanoma and 2.5 % in acral melanoma. It appears that fusion proteins are mutually exclusive for most common oncogenic drivers such as BRAF or NRAS. A further indicator of an increased probability of detecting NTRK-positive tumors could be a low mutation load. Since TRK inhibitors are already available for patients with NTRK fusions, the challenge will be to implement screening for NTRK gene fusions in clinical practice. A possible approach could be to screen BRAF, NRAS and KIT wild-type melanoma patients with next-generation sequencing as soon as they need systemic treatment or at the latest when they have no tumor control on checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Forschner
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Forchhammer
- Department of Dermatology, Histopathology, University Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and neuropathology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
840
|
Solomon JP, Benayed R, Hechtman JF, Ladanyi M. Identifying patients with NTRK fusion cancer. Ann Oncol 2020; 30 Suppl 8:viii16-viii22. [PMID: 31738428 PMCID: PMC6859817 DOI: 10.1093/annonc/mdz384] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Due to the efficacy of tropomyosin receptor kinase (TRK) inhibitor therapy and the recent Food and Drug Administration approval of larotrectinib, it is now clinically important to accurately and efficiently identify patients with neurotrophic TRK (NTRK) fusion-driven cancer. These oncogenic fusions occur when the kinase domain of NTRK1, NTRK2 or NTRK3 fuse with any of a number of N-terminal partners. NTRK fusions are characteristic of a few rare types of cancer, such as secretory carcinoma of the breast or salivary gland and infantile fibrosarcoma, but they are also infrequently seen in some common cancers, such as melanoma, glioma and carcinomas of the thyroid, lung and colon. There are multiple methods for identifying NTRK fusions, including pan-TRK immunohistochemistry, fluorescence in situ hybridisation and sequencing methods, and the advantages and drawbacks of each are reviewed here. While testing algorithms will obviously depend on availability of various testing modalities and economic considerations for each individual laboratory, we propose triaging specimens based on histology and other molecular findings to most efficiently identify tumours harbouring these treatable oncogenic fusions.
Collapse
Affiliation(s)
- J P Solomon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - R Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - M Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
841
|
Yoshino T, Pentheroudakis G, Mishima S, Overman MJ, Yeh KH, Baba E, Naito Y, Calvo F, Saxena A, Chen LT, Takeda M, Cervantes A, Taniguchi H, Yoshida K, Kodera Y, Kitagawa Y, Tabernero J, Burris H, Douillard JY. JSCO-ESMO-ASCO-JSMO-TOS: international expert consensus recommendations for tumour-agnostic treatments in patients with solid tumours with microsatellite instability or NTRK fusions. Ann Oncol 2020; 31:861-872. [PMID: 32272210 DOI: 10.1016/j.annonc.2020.03.299] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/15/2020] [Indexed: 01/05/2023] Open
Abstract
A Japan Society of Clinical Oncology (JSCO)-hosted expert meeting was held in Japan on 27 October 2019, which comprised experts from the JSCO, the Japanese Society of Medical Oncology (JSMO), the European Society for Medical Oncology (ESMO), the American Society of Clinical Oncology (ASCO), and the Taiwan Oncology Society (TOS). The purpose of the meeting was to focus on what we have learnt from both microsatellite instability (MSI)/deficient mismatch repair (dMMR) biomarkers in predicting the efficacy of anti-programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) immunotherapy, and the neurotrophic tyrosine receptor kinase (NTRK) gene fusions in predicting the efficacy of inhibitors of the tropomyosin receptor kinase (TRK) proteins across a range of solid tumour types. The recent regulatory approvals of the anti-PD-1 antibody pembrolizumab and the TRK inhibitors larotrectinib and entrectinib, based on specific tumour biomarkers rather than specific tumour type, have heralded a paradigm shift in cancer treatment approaches. The purpose of the meeting was to develop international expert consensus recommendations on the use of such tumour-agnostic treatments in patients with solid tumours. The aim was to generate a reference document for clinical practice, for pharmaceutical companies in the design of clinical trials, for ethics committees in the approval of clinical trial protocols and for regulatory authorities in relation to drug approvals, with a particular emphasis on diagnostic testing and patient selection.
Collapse
Affiliation(s)
- T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - G Pentheroudakis
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece
| | - S Mishima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - M J Overman
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - K-H Yeh
- Department of Medical Oncology, National Taiwan University Cancer Center and Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - E Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Y Naito
- Department of Experimental Therapeutics/Breast and Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - F Calvo
- Department of Clinical Pharmacology, University of Paris and Institute Gustave Roussy, Villejuif, France
| | - A Saxena
- Department of Medicine, Division of Hematology & Medical Oncology, Thoracic Oncology Service, Weill Cornell Medicine, New York, USA
| | - L-T Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - M Takeda
- Department of Medical Oncology, Kindai University, Osaka, Japan
| | - A Cervantes
- CIBERONC, Department of Medical Oncology, Institute of Health Research, INCLIVIA, University of Valencia, Valencia, Spain
| | - H Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - K Yoshida
- Department of Surgical Oncology, Gifu University, Graduate School of Medicine, Gifu, Japan
| | - Y Kodera
- Department of Gastrointestinal Surgery, Nagoya University, Nagoya, Japan
| | - Y Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - J Tabernero
- Medical Oncology Department, Vall d' Hebron University Hospital, Vall d'Hebron Institute of Oncology (V.H.I.O.), Barcelona, Spain
| | - H Burris
- The Sarah Cannon Research Institute, Nashville, USA
| | | |
Collapse
|
842
|
Faulkner S, Griffin N, Rowe CW, Jobling P, Lombard JM, Oliveira SM, Walker MM, Hondermarck H. Nerve growth factor and its receptor tyrosine kinase TrkA are overexpressed in cervical squamous cell carcinoma. FASEB Bioadv 2020; 2:398-408. [PMID: 32676580 PMCID: PMC7354692 DOI: 10.1096/fba.2020-00016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/02/2020] [Accepted: 05/21/2020] [Indexed: 01/05/2023] Open
Abstract
Nerve growth factor (NGF) and its receptors are increasingly implicated in cancer progression, but their expression in cervical cancer is unclear. The objective of this study was to define the protein expression of NGF, its precursor (proNGF), as well as their receptors, the tyrosine kinase receptor TrkA, the common neurotrophin receptor p75NTR and the pro-neurotrophin receptor sortilin in cervical cancer. Immunohistochemistry was performed in a cohort of cervical cancers (n = 287), including the two major subtypes of the disease: squamous cell carcinomas (SCC) and adenocarcinomas (AC). Normal cervical tissues (n = 28) were also analyzed. Protein expression was determined by computer-based digital quantification of staining intensity and comparative statistical analyses were made with clinicopathological parameters including histological subtype, age, grade, tumor size, lymph node invasion, and stage. The expression of NGF, proNGF, TrkA, p75NTR, and sortilin was higher in cervical cancer compared to normal cervical tissues. NGF and TrkA were found overexpressed in SCC compared to AC (P = .0006 and P < .0001, respectively). The expression of NGF (P = .0053), proNGF (P = .0022), and p75NTR (P = .0002), but not that of TrkA or sortilin, was associated with increasing grade in SCC. In addition, nerve infiltration into the tumor microenvironment was assessed using the pan-neuronal marker PGP9.5. Infiltrating nerves were detected in 27% of cervical tumors and expressed TrkA. Functional investigations using the HELA cervical cancer cell line indicated that the Trk tyrosine kinase inhibitor GNF-5837 reduced cell viability through decreased ERK1/2 activation. Together, these data reveal the overexpression of NGF and TrkA in cervical SCC, suggesting a potential therapeutic value of targeting the NGF-TrkA signaling pathway in this subtype of cervical cancer.
Collapse
Affiliation(s)
- Sam Faulkner
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
| | - Nathan Griffin
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
| | - Christopher W. Rowe
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
- School of Medicine and Public HealthFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Phillip Jobling
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
| | - Janine M. Lombard
- School of Medicine and Public HealthFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- Department of Medical OncologyCalvary Mater NewcastleWaratahNSWAustralia
| | - Sonia M. Oliveira
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
| | - Marjorie M. Walker
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
- School of Medicine and Public HealthFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Hubert Hondermarck
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- Hunter Medical Research InstituteUniversity of NewcastleNew LambtonNSWAustralia
| |
Collapse
|
843
|
Kao YC, Sung YS, Argani P, Swanson D, Alaggio R, Tap W, Wexler L, Dickson BC, Antonescu CR. NTRK3 overexpression in undifferentiated sarcomas with YWHAE and BCOR genetic alterations. Mod Pathol 2020; 33:1341-1349. [PMID: 32034283 PMCID: PMC7329614 DOI: 10.1038/s41379-020-0495-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/25/2020] [Accepted: 01/26/2020] [Indexed: 11/28/2022]
Abstract
The BCOR family of tumors includes a number of undifferentiated sarcomas, occurring in various age groups and anatomic sites, characterized by a spindle and round cell phenotype and diffuse immunoreactivity for BCOR. Prior RNA sequencing data revealed that NTRK3 was a top-upregulated gene in BCOR-CCNB3 sarcomas. In this study, we investigate a large cohort of tumors harboring BCOR/YWHAE genetic alterations for NTRK3 upregulation at both the mRNA and protein levels, compared with other sarcoma types. Pan-Trk immunohistochemistry was assessed for intensity and extent. A correlation between NTRK3 expression and the type of BCOR alteration and BCOR immunoreactivity was also performed. Most soft tissue undifferentiated round cell sarcomas with YWHAE or BCOR rearrangements or BCOR internal tandem duplications (ITD) showed NTRK3, but not NTRK1 or NTRK2, upregulation by RNA sequencing data analysis. Cytoplasmic pan-Trk immunoreactivity was also observed in most soft tissue round cell sarcomas with YWHAE rearrangements (100%), BCOR ITD (80%), and BCOR-CCNB3 fusions (67%), as well as clear cell sarcomas of kidney (75%), another BCOR family tumor, and ossifying fibromyxoid tumors with ZC3H7B-BCOR fusion (100%), with variable staining intensity and extent. Pan-Trk staining was also seen in solitary fibrous tumors (100%) and less frequently in synovial sarcoma and Ewing sarcoma, but rarely in other sarcomas tested. Tumors harboring rare fusion variants of BCOR, such as BCOR-CHD9, a novel fusion identified by targeted RNA sequencing, and KMT2D-BCOR, were also positive for pan-Trk staining and NTRK3 overexpression. In conclusion, NTRK3 upregulation resulting in pan-Trk overexpression is common in the BCOR family of tumors as well as in subsets of BCOR-expressing sarcomas through alternative mechanisms. The therapeutic implication of this finding awaits further investigation.
Collapse
Affiliation(s)
- Yu-Chien Kao
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Shao Sung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Argani
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David Swanson
- Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Rita Alaggio
- Department of Pathology, Bambino Gesu Hospital, Rome, Italy
| | - William Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Leonard Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brendan C. Dickson
- Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | | |
Collapse
|
844
|
Challenges in the Diagnosis of NTRK Fusion-Positive Cancers. J Thorac Oncol 2020; 15:e108-e110. [PMID: 32593446 DOI: 10.1016/j.jtho.2019.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 12/25/2022]
|
845
|
Li Z, Zhou Z, Wu X, Zhou Q, Liao C, Liu Y, Li D, Shen L, Feng D, Yang L. LMP1 promotes nasopharyngeal carcinoma metastasis through NTRK2-mediated anoikis resistance. Am J Cancer Res 2020; 10:2083-2099. [PMID: 32775002 PMCID: PMC7407352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/18/2020] [Indexed: 06/11/2023] Open
Abstract
Anoikis resistance is an important mechanism that mediates tumor metastasis. Studies have found that Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP1) promotes the occurrence, development, and metastasis of nasopharyngeal carcinoma (NPC). However, the related mechanism, especially whether LMP1 is involved in NPC metastasis through anoikis resistance, has not yet been elucidated. In present study, we showed that LMP1 enhanced the ability of NPC cells to resist anoikis by upregulating neurotrophic tyrosine kinase receptor type 2 (NTRK2 or TrkB) expression through NF-κB signaling and promoted the migration and invasion of NPC cells. After knockdown of NTRK2, the p-ERK and p-AKT in NPC cells were inhibited, and twist expression was further reduced, resulting in upregulation of E-cadherin expression and downregulation of vimentin expression. Subsequently, the results of a xenograft experiment showed that inhibiting NTRK2 could reduce LMP1-mediated NPC metastasis in vivo. In summary, these findings demonstrated that EBV-LMP1 upregulates twist expression to promote epithelial-mesenchymal transition (EMT) through the NTRK2-mediated AKT/ERK signaling pathway, thus mediating anoikis resistance and promoting NPC metastasis. These data will provide new molecular markers and potential targets for NPC metastasis.
Collapse
Affiliation(s)
- Zhilan Li
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Zhuan Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South UniversityChangsha, China
| | - Xia Wu
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South UniversityChangsha, China
| | - Qin Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
| | - Chaoliang Liao
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South UniversityChangsha, China
| | - Ying Liu
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Dan Li
- Institue of Molecular Medicine and Oncology, College of Biology, Hunan UniversityChangsha, China
| | - Liangfang Shen
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
| | - Deyun Feng
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Lifang Yang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Xiangya Hospital, Central South UniversityChangsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South UniversityChangsha, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha, China
| |
Collapse
|
846
|
Abstract
For over three decades, a mainstay and goal of clinical oncology has been the development of therapies promoting the effective elimination of cancer cells by apoptosis. This programmed cell death process is mediated by several signalling pathways (referred to as intrinsic and extrinsic) triggered by multiple factors, including cellular stress, DNA damage and immune surveillance. The interaction of apoptosis pathways with other signalling mechanisms can also affect cell death. The clinical translation of effective pro-apoptotic agents involves drug discovery studies (addressing the bioavailability, stability, tumour penetration, toxicity profile in non-malignant tissues, drug interactions and off-target effects) as well as an understanding of tumour biology (including heterogeneity and evolution of resistant clones). While tumour cell death can result in response to therapy, the selection, growth and dissemination of resistant cells can ultimately be fatal. In this Review, we present the main apoptosis pathways and other signalling pathways that interact with them, and discuss actionable molecular targets, therapeutic agents in clinical translation and known mechanisms of resistance to these agents.
Collapse
Affiliation(s)
| | - Wafik S El-Deiry
- The Warren Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
847
|
Alharbi M, Mobark NA, Balbaid AAO, Alanazi FA, Aljabarat WAR, Bakhsh EA, Ramkissoon SH, Abedalthagafi M. Regression of ETV6-NTRK3 Infantile Glioblastoma After First-Line Treatment With Larotrectinib. JCO Precis Oncol 2020; 4:2000017. [PMID: 32923892 PMCID: PMC7446437 DOI: 10.1200/po.20.00017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Musa Alharbi
- Department of Paediatric Oncology, Comprehensive Cancer Centre, King Fahad Medical, Riyadh, Saudi Arabia
| | - Nahla Ali Mobark
- Department of Paediatric Oncology, Comprehensive Cancer Centre, King Fahad Medical, Riyadh, Saudi Arabia
| | - Ali Abdullah O Balbaid
- Radiation Oncology Department, Comprehensive Cancer Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fatmah A Alanazi
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Eman A Bakhsh
- Radiology Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Shakti H Ramkissoon
- Foundation Medicine, Morrisville, NC.,Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Malak Abedalthagafi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| |
Collapse
|
848
|
Serrano C, George S. Gastrointestinal Stromal Tumor: Challenges and Opportunities for a New Decade. Clin Cancer Res 2020; 26:5078-5085. [PMID: 32601076 DOI: 10.1158/1078-0432.ccr-20-1706] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/26/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Gastrointestinal stromal tumor (GIST) provides a paradigm to evaluate new molecularly targeted therapies and to identify structural and functional mechanisms for drug response and resistance. Drug development in GIST has successfully exploited the high reliance on KIT/PDGFRA oncogenic signaling as a therapeutic vulnerability. The recent arrival of avapritinib and ripretinib to the GIST arena has aimed to further improve on precision kinase inhibition and address tumor heterogeneity in imatinib-resistant GIST. The two main clinical challenges for the forthcoming years entail tumor eradication in patients with early-stage GIST, and maximization of tumor response in late-stage disease. To succeed, we will need to better understand the mechanisms behind adaptation to KIT inhibition and apoptosis evasion, tumor evolution after successive lines of treatment, and to explore clinically novel creative therapeutic strategies, with the overarching goal to tackle the intrinsic oncogenic complexity while minimizing adverse events.
Collapse
Affiliation(s)
- César Serrano
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology, Barcelona, Spain. .,Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Suzanne George
- Department of Medical Oncology, Sarcoma Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
849
|
Tischoff I, Tannapfel A. [CUP in the liver]. DER PATHOLOGE 2020; 41:495-504. [PMID: 32588138 DOI: 10.1007/s00292-020-00803-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Hepatic involvement is one of the most common manifestations in cancer of unknown primary (CUP) syndrome. The most frequent secondary neoplasms of the liver are carcinomas and malignant melanomas. Most common carcinoma metastases are adenocarcinomas originating from the digestive system or metastases of breast and lung carcinomas. Therefore, hepatic CUP syndrome is an exclusion diagnosis. Immunohistochemistry and molecular examinations are an important part of histopathological diagnosis. They do not only serve to identify the tissue of histologically origin or possible primary tumor, but also contribute to the selection of a personalized targeted therapy by detecting so-called druggable targets in the interdisciplinary management.
Collapse
Affiliation(s)
- Iris Tischoff
- Institut für Pathologie,an den Universitätskliniken Bergmannsheil, Ruhr-Universität Bochum, Bürkle de la Camp-Platz 1, 44789, Bochum, Deutschland.
| | - Andrea Tannapfel
- Institut für Pathologie,an den Universitätskliniken Bergmannsheil, Ruhr-Universität Bochum, Bürkle de la Camp-Platz 1, 44789, Bochum, Deutschland
| |
Collapse
|
850
|
U.S. Phase I First-in-human Study of Taletrectinib (DS-6051b/AB-106), a ROS1/TRK Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res 2020; 26:4785-4794. [DOI: 10.1158/1078-0432.ccr-20-1630] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/06/2020] [Accepted: 06/24/2020] [Indexed: 11/16/2022]
|