101
|
Bokemeyer C, Paracha N, Lassen U, Italiano A, Sullivan SD, Marian M, Brega N, Garcia-Foncillas J. Survival Outcomes of Patients With Tropomyosin Receptor Kinase Fusion-Positive Cancer Receiving Larotrectinib Versus Standard of Care: A Matching-Adjusted Indirect Comparison Using Real-World Data. JCO Precis Oncol 2023; 7:e2200436. [PMID: 36689698 PMCID: PMC9928633 DOI: 10.1200/po.22.00436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Larotrectinib, a highly specific tropomyosin receptor kinase (TRK) inhibitor, previously demonstrated high response rates in single-arm trials of patients with TRK fusion-positive cancer, but there are limited data on comparative effectiveness against standard-of-care (SoC) regimens used in routine health care practice, before widespread adoption of TRK inhibitors as SoC for TRK fusion-positive cancers. Matching-adjusted indirect comparison, a validated methodology that balances population characteristics to facilitate cross-trial comparisons, was used to compare the overall survival (OS) of larotrectinib versus non-TRK-inhibitor SoC. MATERIALS AND METHODS Individual patient data from three larotrectinib trials (ClinicalTrials.gov identifiers: NCT02122913, NCT02637687, and NCT02576431) were compared with published aggregate real-world data from patients with locally advanced/metastatic TRK fusion-positive cancer identified in the Flatiron Health/Foundation Medicine database. OS was defined as the time from advanced/metastatic disease diagnosis to death. After matching population characteristics, the analyses included (1) a log-rank test of equality to test whether the two groups were similar before larotrectinib initiation; and (2) estimation of treatment effect of larotrectinib versus non-TRK-inhibitor SoC. These analyses are limited to prognostic variables available in real-world data. RESULTS Eighty-five larotrectinib patients and 28 non-TRK-inhibitor SoC patients were included in the analyses. After matching, log-rank testing showed no difference in baseline characteristics between the two groups (P = .31). After matching, larotrectinib was associated with a 78% lower risk of death, compared with non-TRK-inhibitor SoC (adjusted hazard ratio, 0.22 [95% CI, 0.09 to 0.52]; P = .001); median OS was 39.7 months (95% CI: 16.4, NE [not estimable]) for larotrectinib and 10.2 months (95% CI: 7.2, 14.1) for SoC. CONCLUSION Matching-adjusted indirect comparison analyses suggest longer OS with larotrectinib, compared with non-TRK-inhibitor SoC, in adult patients with TRK fusion-positive cancer.
Collapse
Affiliation(s)
- Carsten Bokemeyer
- University Medical Centre Hamburg Eppendorf, Hamburg, Germany,Carsten Bokemeyer, MD, Department Oncology, Hematology and BMT with Section of Pneumology, Universitaetsklinikum Hamburg—Eppendorf, Martinistrasse 52, D 20246 Hamburg, Germany; e-mail:
| | | | | | | | - Sean D. Sullivan
- CHOICE Institute, School of Pharmacy, University of Washington, Seattle, WA
| | | | | | - Jesus Garcia-Foncillas
- University Cancer Institute and the Department of Oncology, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid, Spain
| |
Collapse
|
102
|
Vingiani A, Lorenzini D, Conca E, Volpi CC, Trupia DV, Gloghini A, Perrone F, Tamborini E, Dagrada GP, Agnelli L, Capone I, Busico A, Pruneri G. Pan-TRK immunohistochemistry as screening tool for NTRK fusions: A diagnostic workflow for the identification of positive patients in clinical practice. Cancer Biomark 2023; 38:301-309. [PMID: 37545217 DOI: 10.3233/cbm-220357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND Pan-TRK inhibitors Entrectinib and Larotrectinib have been recently approved as tumor-agnostic therapies in NTRK1-2-3 rearranged patients and there is therefore an urgent need to identify reliable and accessible biomarkers for capturing NTRK fusions in the real-world practice. OBJECTIVE We aim to assess the analytical validity of the recently released pan-TRK assay (Ventana), running a head-to-head comparison between immunohistochemistry and Archer FusionPlex Lung Panel (ArcherDX) that is designed to detect key fusions in 13 genes, also including NTRK1-3. METHODS Pan-TRK IHC and NGS analysis were conducted on a retrospective/prospective cohort of 124 cancer patients (carcinomas, 93 cases; soft tissue sarcomas, 19; primary central nervous system tumours, 10; and neuroblastomas, 2). FISH data were available in most of the IHC/NGS discordant cases. RESULTS A comparison between IHC and NGS results was carried out in 117 cases: among 30 pan-TRK positive cases, NTRK rearrangement by NGS was found in 11 (37%), while one of the 87 (1.1%) pan-TRK negative cases (a case of NSCLC) showed a TPM3-NRTK1 rearrangement by NGS. Accordingly, sensitivity and specificity of IHC in predicting NTRK status were 91.7% and 81.9%, respectively, while negative (NPV) and positive predictive value (PPV) were 98.8% and 36.7%, respectively. CONCLUSIONS These data lead to suggest that IHC with VENTANA pan-TRK antibody can be a reliable screening tool for the identification of patients potentially bearing NTRK rearranged tumours.
Collapse
Affiliation(s)
- Andrea Vingiani
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Daniele Lorenzini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Elena Conca
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Chiara Costanza Volpi
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Desirè Viola Trupia
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Federica Perrone
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Elena Tamborini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Gian Paolo Dagrada
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Luca Agnelli
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Iolanda Capone
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Adele Busico
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
| | - Giancarlo Pruneri
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
| |
Collapse
|
103
|
Riedel R, Fassunke J, Tumbrink HL, Scheel AH, Heydt C, Hieggelke L, Scheffler M, Heimsoeth A, Nogova L, Michels S, Weber JP, Fischer RN, Eisert A, Westphal T, Schaufler D, Siemanowski J, Ihle MA, Wagener-Ryczek S, Castiglione R, Pappesch R, Rehker J, Jürgens J, Stoelben E, Bunck A, Kobe C, Merkelbach-Bruse S, Sos ML, Büttner R, Wolf J. Resistance to MET inhibition in MET-dependent NSCLC and therapeutic activity after switching from type I to type II MET inhibitors. Eur J Cancer 2023; 179:124-135. [PMID: 36521334 DOI: 10.1016/j.ejca.2022.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Resistance to MET inhibition occurs inevitably in MET-dependent non-small cell lung cancer and the underlying mechanisms are insufficiently understood. We describe resistance mechanisms in patients with MET exon 14 skipping mutation (METΔex14), MET amplification, and MET fusion and report treatment outcomes after switching therapy from type I to type II MET inhibitors. MATERIALS AND METHODS Pre- and post-treatment biopsies were analysed by NGS (next generation sequencing), digital droplet PCR (polymerase chain reaction), and FISH (fluorescense in situ hybridization). A patient-derived xenograft model was generated in one case. RESULTS Of 26 patients with MET tyrosine kinase inhibitor treatment, eight had paired pre- and post-treatment biopsies (Three with MET amplification, three with METΔex14, two with MET fusions (KIF5B-MET and PRKAR2B-MET).) In six patients, mechanisms of resistance were detected, whereas in two cases, the cause of resistance remained unclear. We found off-target resistance mechanisms in four cases with KRAS mutations and HER2 amplifications appearing. Two patients exhibited second-site MET mutations (p.D1246N and p. Y1248H). Three patients received type I and type II MET tyrosine kinase inhibitors sequentially. In two cases, further progressive disease was seen hereafter. The patient with KIF5B-MET fusion received three different MET inhibitors and showed long-lasting stable disease and a repeated response after switching therapy, respectively. CONCLUSION Resistance to MET inhibition is heterogeneous with on- and off-target mechanisms occurring regardless of the initial MET aberration. Switching therapy between different types of kinase inhibitors can lead to repeated responses in cases with second-site mutations. Controlled clinical trials in this setting with larger patient numbers are needed, as evidence to date is limited to preclinical data and case series.
Collapse
Affiliation(s)
- Richard Riedel
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Jana Fassunke
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Hannah L Tumbrink
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Germany
| | - Andreas H Scheel
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Carina Heydt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Lena Hieggelke
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Matthias Scheffler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Alena Heimsoeth
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Germany
| | - Lucia Nogova
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Sebastian Michels
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Jan-Phillip Weber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Rieke N Fischer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Anna Eisert
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Theresa Westphal
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Diana Schaufler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany
| | - Janna Siemanowski
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Michaela A Ihle
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Svenja Wagener-Ryczek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | | | - Roberto Pappesch
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Jan Rehker
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Jessica Jürgens
- Lung Clinic Merheim, Hospital of the City of Cologne, University of Witten-Herdecke, Germany
| | - Erich Stoelben
- Lung Clinic Merheim, Hospital of the City of Cologne, University of Witten-Herdecke, Germany
| | - Anne Bunck
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Radiology, Germany
| | - Carsten Kobe
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Nuclear Medicine, Germany
| | - Sabine Merkelbach-Bruse
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany
| | - Martin L Sos
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Reinhard Büttner
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department of Pathology, Molecular Pathology, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Jürgen Wolf
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology, Department I of Internal Medicine, Germany; Lung Cancer Group, Cologne, Germany.
| |
Collapse
|
104
|
De Faria FW, Schieffer KM, Pierson CR, Boue DR, LaHaye S, Miller KE, Amayiri N, Koboldt DC, Lichtenberg T, Leraas K, Brennan P, Kelly B, White P, Magrini V, Wilson RK, Mardis ER, Cottrell CE, Rusin J, Finlay JL, Osorio DS. Infantile metastatic ependymoma with a novel molecular profile and favorable outcome to intensive chemotherapy without irradiation: Case-based review. Genes Chromosomes Cancer 2023; 62:39-46. [PMID: 35716171 DOI: 10.1002/gcc.23081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/06/2022] Open
Abstract
Ependymal tumors are the third most common brain tumor under 14 years old. Even though metastatic disease is a rare event, it affects mostly young children and carries an adverse prognosis. The factors associated with dissemination and the best treatment approach have not yet been established and there is limited published data on how to manage metastatic disease, especially in patients under 3 years of age. We provide a review of the literature on clinical characteristics and radiation-sparing treatments for metastatic ependymoma in children under 3 years of age treated. The majority (73%) of the identified cases were above 12 months old and had the PF as the primary site at diagnosis. Chemotherapy-based approaches, in different regimens, were used with radiation reserved for progression or relapse. The prognosis varied among the studies, with an average of 50%-58% overall survival. This study also describes the case of a 7-month-old boy with metastatic posterior fossa (PF) ependymoma, for whom we identified a novel SPECC1L-RAF1 gene fusion using a patient-centric comprehensive molecular profiling protocol. The patient was successfully treated with intensive induction chemotherapy followed by high-dose chemotherapy and autologous hematopoietic progenitor cell rescue (AuHSCR). Currently, the patient is in continuous remission 5 years after his diagnosis, without radiation therapy. The understanding of the available therapeutic approaches may assist physicians in their management of such patients. This report also opens the perspective of newly identified molecular alterations in metastatic ependymomas that might drive more chemo-sensitive tumors.
Collapse
Affiliation(s)
- Flavia Watusi De Faria
- Department of Pediatric Hematology and Oncology, Hospital da Criança de Brasilia, Brasilia, Distrito Federal, Brazil.,Department of Pediatric Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Kathleen M Schieffer
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Christopher R Pierson
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pathology, The Ohio State University, Columbus, Ohio, USA.,The Department of Biomedical Education and Anatomy, Division of Anatomy, The Ohio State University, Columbus, Ohio, USA
| | - Daniel R Boue
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie LaHaye
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Katherine E Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Nisreen Amayiri
- Division of Pediatric Hematology/Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Daniel C Koboldt
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Tara Lichtenberg
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kristen Leraas
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Patrick Brennan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Ben Kelly
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Peter White
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Vincent Magrini
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Richard K Wilson
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Elaine R Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Catherine E Cottrell
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA.,Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jerome Rusin
- Department of Radiology, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, USA
| | - Jonathan L Finlay
- The Division of Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, USA
| | - Diana S Osorio
- The Division of Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
105
|
Abstract
INTRODUCTION Neurotrophic tyrosine receptor kinase (NTRK) gene fusions occur in ~ 0.3% of all solid tumours but are enriched in some rare tumour types. Tropomyosin receptor kinase (TRK) inhibitors larotrectinib and entrectinib are approved as tumour-agnostic therapies for solid tumours harbouring NTRK fusions. METHODS This study investigated the prevalence of NTRK fusions in Canadian patients and also aimed to help guide NTRK testing paradigms through analysis of data reported from a national clinical diagnostic testing program between September 2019 and July 2021. RESULTS Of 1,687 patients included in the final analysis, NTRK fusions were detected in 0.71% (n = 12) of patients representing salivary gland carcinoma (n = 3), soft tissue sarcoma (n = 3), CNS (n = 3), and one in each of melanoma, lung, and colorectal cancer. All three salivary gland carcinomas contained ETV6-NTRK3 fusions. Thirteen (0.77%) clinically actionable incidental findings were also detected. Two of the 13 samples containing incidental findings were NTRK fusion-positive (GFOD1-NTRK2, FGFR3-TACC3 in a glioblastoma and AFAP1-NTRK2, BRAF c.1799T>A in a glioma). The testing algorithm screened most patient samples via pan-TRK immunohistochemistry (IHC), whereas samples from the central nervous system (CNS), pathognomonic cancers, and confirmed/ putative NTRK fusion-positive samples identified under research protocols were reflexed straight to next-generation sequencing (NGS). CONCLUSION These findings highlight the benefit and practicality of a diagnostic testing program to identify patients suitable for tumour-agnostic TRK inhibitor therapies, as well as other targeted therapies, due to clinically actionable incidental findings identified. Collectively, these findings may inform future guidance on selecting the appropriate testing approach per tumour type and on optimal NTRK testing algorithms.
Collapse
|
106
|
Sorokin M, Rabushko E, Rozenberg JM, Mohammad T, Seryakov A, Sekacheva M, Buzdin A. Clinically relevant fusion oncogenes: detection and practical implications. Ther Adv Med Oncol 2022; 14:17588359221144108. [PMID: 36601633 PMCID: PMC9806411 DOI: 10.1177/17588359221144108] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/22/2022] [Indexed: 12/28/2022] Open
Abstract
Mechanistically, chimeric genes result from DNA rearrangements and include parts of preexisting normal genes combined at the genomic junction site. Some rearranged genes encode pathological proteins with altered molecular functions. Those which can aberrantly promote carcinogenesis are called fusion oncogenes. Their formation is not a rare event in human cancers, and many of them were documented in numerous study reports and in specific databases. They may have various molecular peculiarities like increased stability of an oncogenic part, self-activation of tyrosine kinase receptor moiety, and altered transcriptional regulation activities. Currently, tens of low molecular mass inhibitors are approved in cancers as the drugs targeting receptor tyrosine kinase (RTK) oncogenic fusion proteins, that is, including ALK, ABL, EGFR, FGFR1-3, NTRK1-3, MET, RET, ROS1 moieties. Therein, the presence of the respective RTK fusion in the cancer genome is the diagnostic biomarker for drug prescription. However, identification of such fusion oncogenes is challenging as the breakpoint may arise in multiple sites within the gene, and the exact fusion partner is generally unknown. There is no gold standard method for RTK fusion detection, and many alternative experimental techniques are employed nowadays to solve this issue. Among them, RNA-seq-based methods offer an advantage of unbiased high-throughput analysis of only transcribed RTK fusion genes, and of simultaneous finding both fusion partners in a single RNA-seq read. Here we focus on current knowledge of biology and clinical aspects of RTK fusion genes, related databases, and laboratory detection methods.
Collapse
Affiliation(s)
| | - Elizaveta Rabushko
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia,I.M. Sechenov First Moscow State Medical
University, Moscow, Russia
| | | | - Tharaa Mohammad
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia
| | | | - Marina Sekacheva
- I.M. Sechenov First Moscow State Medical
University, Moscow, Russia
| | - Anton Buzdin
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia,I.M. Sechenov First Moscow State Medical
University, Moscow, Russia,Shemyakin-Ovchinnikov Institute of Bioorganic
Chemistry, Moscow, Russia,PathoBiology Group, European Organization for
Research and Treatment of Cancer (EORTC), Brussels, Belgium
| |
Collapse
|
107
|
Li Q, Li Z, Luo T, Shi H. Targeting the PI3K/AKT/mTOR and RAF/MEK/ERK pathways for cancer therapy. MOLECULAR BIOMEDICINE 2022; 3:47. [PMID: 36539659 PMCID: PMC9768098 DOI: 10.1186/s43556-022-00110-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022] Open
Abstract
The PI3K/AKT/mTOR and RAF/MEK/ERK pathways are commonly activated by mutations and chromosomal translocation in vital targets. The PI3K/AKT/mTOR signaling pathway is dysregulated in nearly all kinds of neoplasms, with the component in this pathway alternations. RAF/MEK/ERK signaling cascades are used to conduct signaling from the cell surface to the nucleus to mediate gene expression, cell cycle processes and apoptosis. RAS, B-Raf, PI3K, and PTEN are frequent upstream alternative sites. These mutations resulted in activated cell growth and downregulated cell apoptosis. The two pathways interact with each other to participate in tumorigenesis. PTEN alterations suppress RAF/MEK/ERK pathway activity via AKT phosphorylation and RAS inhibition. Several inhibitors targeting major components of these two pathways have been supported by the FDA. Dozens of agents in these two pathways have attracted great attention and have been assessed in clinical trials. The combination of small molecular inhibitors with traditional regimens has also been explored. Furthermore, dual inhibitors provide new insight into antitumor activity. This review will further comprehensively describe the genetic alterations in normal patients and tumor patients and discuss the role of targeted inhibitors in malignant neoplasm therapy. We hope this review will promote a comprehensive understanding of the role of the PI3K/AKT/mTOR and RAF/MEK/ERK signaling pathways in facilitating tumors and will help direct drug selection for tumor therapy.
Collapse
Affiliation(s)
- Qingfang Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, China
| | - Zhihui Li
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, PR China
| | - Ting Luo
- Department of Breast, Cancer Center, West China Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| |
Collapse
|
108
|
Li Y, Wei C, Wang W, Li Q, Wang Z. Tropomyosin receptor kinase B (TrkB) signalling: targeted therapy in neurogenic tumours. J Pathol Clin Res 2022; 9:89-99. [PMID: 36533776 PMCID: PMC9896160 DOI: 10.1002/cjp2.307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Tropomyosin receptor kinase B (TrkB), a transmembrane receptor protein, has been found to play a pivotal role in neural development. This protein is encoded by the neurotrophic receptor tyrosine kinase 2 (NTRK2) gene, and its abnormal activation caused by NTRK2 overexpression or fusion can contribute to tumour initiation, progression, and resistance to therapeutics in multiple types of neurogenic tumours. Targeted therapies for this mechanism have been designed and developed in preclinical and clinical studies, including selective TrkB inhibitors and pan-TRK inhibitors. This review describes the gene structure, biological function, abnormal TrkB activation mechanism, and current-related targeted therapies in neurogenic tumours.
Collapse
Affiliation(s)
- Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Zhi‐Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
109
|
A narrative review of cancer molecular diagnostics: past, present, and future. JOURNAL OF BIO-X RESEARCH 2022. [DOI: 10.1097/jbr.0000000000000136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
110
|
Wang Z, Xing Y, Li B, Li X, Liu B, Wang Y. Molecular pathways, resistance mechanisms and targeted interventions in non-small-cell lung cancer. MOLECULAR BIOMEDICINE 2022; 3:42. [PMID: 36508072 PMCID: PMC9743956 DOI: 10.1186/s43556-022-00107-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/03/2022] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. The discovery of tyrosine kinase inhibitors effectively targeting EGFR mutations in lung cancer patients in 2004 represented the beginning of the precision medicine era for this refractory disease. This great progress benefits from the identification of driver gene mutations, and after that, conventional and new technologies such as NGS further illustrated part of the complex molecular pathways of NSCLC. More targetable driver gene mutation identification in NSCLC patients greatly promoted the development of targeted therapy and provided great help for patient outcomes including significantly improved survival time and quality of life. Herein, we review the literature and ongoing clinical trials of NSCLC targeted therapy to address the molecular pathways and targeted intervention progress in NSCLC. In addition, the mutations in EGFR gene, ALK rearrangements, and KRAS mutations in the main sections, and the less common molecular alterations in MET, HER2, BRAF, ROS1, RET, and NTRK are discussed. The main resistance mechanisms of each targeted oncogene are highlighted to demonstrate the current dilemma of targeted therapy in NSCLC. Moreover, we discuss potential therapies to overcome the challenges of drug resistance. In this review, we manage to display the current landscape of targetable therapeutic patterns in NSCLC in this era of precision medicine.
Collapse
Affiliation(s)
- Zixi Wang
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Yurou Xing
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Bingjie Li
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Xiaoyu Li
- grid.412901.f0000 0004 1770 1022Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan China ,grid.412901.f0000 0004 1770 1022State Key Laboratory Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Bin Liu
- grid.54549.390000 0004 0369 4060Department of Medical Oncology, School of Medicine, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, Sichuan China
| | - Yongsheng Wang
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China ,grid.412901.f0000 0004 1770 1022State Key Laboratory Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| |
Collapse
|
111
|
Zhang Y, Chan S, He R, Liu Y, Song X, Tu ZC, Ren X, Zhou Y, Zhang Z, Wang Z, Zhou F, Ding K. 1-Methyl-3-((4-(quinolin-4-yloxy)phenyl)amino)-1H-pyrazole-4-carboxamide derivatives as new rearranged during Transfection (RET) kinase inhibitors capable of suppressing resistant mutants in solvent-front regions. Eur J Med Chem 2022; 244:114862. [DOI: 10.1016/j.ejmech.2022.114862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/28/2022]
|
112
|
Zhao W, Sun X, Shi L, Cai SZ, Ma ZR. Discovery of novel analogs of KHS101 as transforming acidic coiled coil containing protein 3 (TACC3) inhibitors for the treatment of glioblastoma. Eur J Med Chem 2022; 244:114874. [DOI: 10.1016/j.ejmech.2022.114874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022]
|
113
|
Moran JMT, Le LP, Nardi V, Golas J, Farahani AA, Signorelli S, Onozato ML, Foreman RK, Duncan LM, Lawrence DP, Lennerz JK, Dias-Santagata D, Hoang MP. Identification of fusions with potential clinical significance in melanoma. Mod Pathol 2022; 35:1837-1847. [PMID: 35871080 DOI: 10.1038/s41379-022-01138-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022]
Abstract
Though uncommon in melanoma, gene fusions may have therapeutic implications. Next generation sequencing-based clinical assays, designed to detect relevant gene fusions, mutations, and copy number changes, were performed on 750 melanomas (375 primary and 375 metastases) at our institution from 2014-2021. These included 599 (80%) cutaneous, 38 (5%) acral, 11 (1.5%) anorectal, 23 (3%) sinonasal, 27 (3.6%) eye (uveal/ conjunctiva), 11 (1.5%) genital (vulva/penile), and 41 (5.5%) melanomas of unknown primary. Sixteen fusions (2%) were detected in samples from 16 patients: 12/599 (2%) cutaneous, 2/38 (5%) acral, 1/9 (11%) vulva, 1/23(4.3%) sinonasal; and 12/16 (75%) fusions were potentially targetable. We identified two novel rearrangements: NAGS::MAST2 and NOTCH1::GNB1; and two fusions that have been reported in other malignancies but not in melanoma: CANT1::ETV4 (prostate cancer) and CCDC6::RET (thyroid cancer). Additional fusions, previously reported in melanoma, included: EML4::ALK, MLPH::ALK, AGAP3::BRAF, AGK::BRAF, CDH3::BRAF, CCT8::BRAF, DIP2B::BRAF, EFNB1::RAF1, LRCH3::RAF1, MAP4::RAF1, RUFY1::RAF1, and ADCY2::TERT. Fusion positive melanomas harbored recurrent alterations in TERT and CDKN2A, among others. Gene fusions were exceedingly rare (0.2%) in BRAF/RAS/NF1-mutant tumors and were detected in 5.6% of triple wild-type melanomas. Interestingly, gene rearrangements were significantly enriched within the subset of triple wild-type melanomas that harbor TERT promoter mutations (18% versus 2%, p < 0.0001). Thirteen (81%) patients were treated with immunotherapy for metastatic disease or in the adjuvant setting. Six of 12 (50%) patients with potentially actionable fusions progressed on immunotherapy, and 3/6 (50%) were treated with targeted agents (ALK and MEK inhibitors), 2 off-label and 1 as part of a clinical trial. One patient with an AGAP3::BRAF fusion positive melanoma experienced a 30-month long response to trametinib. We show that, detecting fusions, especially in triple wild-type melanomas with TERT promoter mutations, may have a clinically significant impact in patients with advanced disease who have failed front-line immunotherapy.
Collapse
Affiliation(s)
- Jakob M T Moran
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Long P Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Josephine Golas
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander A Farahani
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylvia Signorelli
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maristela L Onozato
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruth K Foreman
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lyn M Duncan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donald P Lawrence
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jochen K Lennerz
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Mai P Hoang
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
114
|
Levillayer L, Cassonnet P, Declercq M, Santos MD, Lebreton L, Danezi K, Demeret C, Sakuntabhai A, Jacob Y, Bureau JF. SKAP2 Modular Organization Differently Recognizes SRC Kinases Depending on Their Activation Status and Localization. Mol Cell Proteomics 2022; 22:100451. [PMID: 36423812 PMCID: PMC9792355 DOI: 10.1016/j.mcpro.2022.100451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/12/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Dimerization of SRC kinase adaptor phosphoprotein 2 (SKAP2) induces an increase of binding for most SRC kinases suggesting a fine-tuning with transphosphorylation for kinase activation. This work addresses the molecular basis of SKAP2-mediated SRC kinase regulation through the lens of their interaction capacities. By combining a luciferase complementation assay and extensive site-directed mutagenesis, we demonstrated that SKAP2 interacts with SRC kinases through a modular organization depending both on their phosphorylation-dependent activation and subcellular localization. SKAP2 contains three interacting modules consisting in the dimerization domain, the SRC homology 3 (SH3) domain, and the second interdomain located between the Pleckstrin homology and the SH3 domains. Functionally, the dimerization domain is necessary and sufficient to bind to most activated and myristyl SRC kinases. In contrast, the three modules are necessary to bind SRC kinases at their steady state. The Pleckstrin homology and SH3 domains of SKAP2 as well as tyrosines located in the interdomains modulate these interactions. Analysis of mutants of the SRC kinase family member hematopoietic cell kinase supports this model and shows the role of two residues, Y390 and K7, on its degradation following activation. In this article, we show that a modular architecture of SKAP2 drives its interaction with SRC kinases, with the binding capacity of each module depending on both their localization and phosphorylation state activation. This work opens new perspectives on the molecular mechanisms of SRC kinases activation, which could have significant therapeutic impact.
Collapse
Affiliation(s)
- Laurine Levillayer
- Unité de Génétique Fonctionnelle des Maladies Infectieuses (GFMI), CNRS UMR 2000, Institut Pasteur, Université de Paris, Paris, France
| | - Patricia Cassonnet
- Unité de Génétique Moléculaire des Virus à ARN (GMVR), CNRS UMR3569, Institut Pasteur, Université de Paris, Paris, France
| | - Marion Declercq
- Unité de Génétique Moléculaire des Virus à ARN (GMVR), CNRS UMR3569, Institut Pasteur, Université de Paris, Paris, France
| | - Mélanie Dos Santos
- Unité de Génétique Moléculaire des Virus à ARN (GMVR), CNRS UMR3569, Institut Pasteur, Université de Paris, Paris, France
| | - Louis Lebreton
- Unité de Génétique Fonctionnelle des Maladies Infectieuses (GFMI), CNRS UMR 2000, Institut Pasteur, Université de Paris, Paris, France
| | - Katerina Danezi
- Unité de Génétique Fonctionnelle des Maladies Infectieuses (GFMI), CNRS UMR 2000, Institut Pasteur, Université de Paris, Paris, France
| | - Caroline Demeret
- Unité de Génétique Moléculaire des Virus à ARN (GMVR), CNRS UMR3569, Institut Pasteur, Université de Paris, Paris, France
| | - Anavaj Sakuntabhai
- Unité de Génétique Fonctionnelle des Maladies Infectieuses (GFMI), CNRS UMR 2000, Institut Pasteur, Université de Paris, Paris, France
| | - Yves Jacob
- Unité de Génétique Moléculaire des Virus à ARN (GMVR), CNRS UMR3569, Institut Pasteur, Université de Paris, Paris, France
| | - Jean-François Bureau
- Unité de Génétique Fonctionnelle des Maladies Infectieuses (GFMI), CNRS UMR 2000, Institut Pasteur, Université de Paris, Paris, France,For correspondence: Jean-François Bureau
| |
Collapse
|
115
|
Wang T, Wei L, Lu Q, Shao Y, You S, Yin JC, Wang S, Shao Y, Chen Z, Wang Z. Landscape of potentially targetable receptor tyrosine kinase fusions in diverse cancers by DNA-based profiling. NPJ Precis Oncol 2022; 6:84. [DOI: 10.1038/s41698-022-00325-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022] Open
Abstract
AbstractRecurrent fusions of receptor tyrosine kinases (RTKs) are often driving events in tumorigenesis that carry important diagnostic value and are potentially targetable by the increasing number of tyrosine kinase inhibitors (TKIs). Here, we characterized the spectrum of 1324 RTK fusions with intact kinase domains in solid tumors by DNA-based high-throughput sequencing. Overall, the prevalence of RTK fusions were 4.7%, with variable frequencies and diverse genomic structures and fusion partners across cancer types. Cancer types, such as thyroid cancers, urological cancers and neuroendocrine tumors are selective in the RTK fusions they carry, while others exhibit highly complex spectra of fusion events. Notably, most RTKs were promiscuous in terms of the partner genes they recombine with. A large proportion of RTK fusions had one of the breakpoints localized to intergenic regions. Comprehensive genomic profiling revealed differences in co-mutational patterns pre- and post-TKI treatments across various RTK fusions. At baseline, multiple cases were detected with co-occurring RTK fusions or concomitant oncogenic mutations in driver genes, such as KRAS and EGFR. Following TKI resistance, we observed differences in potential on- and off-target resistance mutations among fusion variants. For example, the EML4-ALK v3 variant displayed more complex on-target resistance mechanisms, which might explain the reduced survival outcome compared with the v1 variant. Finally, we identified two lung cancer patients with MET+ and NTRK1+ tumors, respectively, who responded well to crizotinib treatment. Taken together, our findings demonstrate the diagnostic and prognostic values of screening for RTK fusions using DNA-based sequencing in solid tumors.
Collapse
|
116
|
PANAGOPOULOS IOANNIS, HEIM SVERRE. Neoplasia-associated Chromosome Translocations Resulting in Gene Truncation. Cancer Genomics Proteomics 2022; 19:647-672. [PMID: 36316036 PMCID: PMC9620447 DOI: 10.21873/cgp.20349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/27/2022] Open
Abstract
Chromosomal translocations in cancer as well as benign neoplasias typically lead to the formation of fusion genes. Such genes may encode chimeric proteins when two protein-coding regions fuse in-frame, or they may result in deregulation of genes via promoter swapping or translocation of the gene into the vicinity of a highly active regulatory element. A less studied consequence of chromosomal translocations is the fusion of two breakpoint genes resulting in an out-of-frame chimera. The breaks then occur in one or both protein-coding regions forming a stop codon in the chimeric transcript shortly after the fusion point. Though the latter genetic events and mechanisms at first awoke little research interest, careful investigations have established them as neither rare nor inconsequential. In the present work, we review and discuss the truncation of genes in neoplastic cells resulting from chromosomal rearrangements, especially from seemingly balanced translocations.
Collapse
Affiliation(s)
- IOANNIS PANAGOPOULOS
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - SVERRE HEIM
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
117
|
Chłopek M, Lasota J, Thompson LDR, Szczepaniak M, Kuźniacka A, Hińcza K, Kubicka K, Kaczorowski M, Newford M, Liu Y, Agaimy A, Biernat W, Durzyńska M, Dziuba I, Hartmann A, Inaguma S, Iżycka-Świeszewska E, Kato H, Kopczyński J, Michal M, Michal M, Pęksa R, Prochorec-Sobieszek M, Starzyńska A, Takahashi S, Wasąg B, Kowalik A, Miettinen M. Alterations in key signaling pathways in sinonasal tract melanoma. A molecular genetics and immunohistochemical study of 90 cases and comprehensive review of the literature. Mod Pathol 2022; 35:1609-1617. [PMID: 35978013 DOI: 10.1038/s41379-022-01122-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/09/2022]
Abstract
Sinonasal mucosal melanoma is a rare tumor arising within the nasal cavity, paranasal sinuses, or nasopharynx (sinonasal tract). This study evaluated 90 cases diagnosed in 29 males and 61 females with median age 68 years. Most tumors involved the nasal cavity and had an epithelioid morphology. Spectrum of research techniques used in this analysis includes targeted-DNA and -RNA next-generation sequencing, Sanger sequencing, fluorescence in situ hybridization and immunohistochemistry. Sinonasal melanomas were commonly driven by RAS (38/90, 42%), especially NRAS (n = 36) mutations and rarely (4/90, 4%) displayed BRAF pathogenic variants. BRAF/RAS mutants were more frequent among paranasal sinuses (10/14, 71%) than nasal (26/64, 41%) tumors. BRAF/RAS-wild type tumors occasionally harbored alterations of the key components and regulators of Ras-MAPK signaling pathway: NF1 mutations (1/17, 6%) or NF1 locus deletions (1/25, 4%), SPRED1 (3/25, 12%), PIK3CA (3/50, 6%), PTEN (4/50, 8%) and mTOR (1/50, 2%) mutations. These mutations often occurred in a mutually exclusive manner. In several tumors some of which were NRAS mutants, TP53 was deleted (6/48, 13%) and/or mutated (5/90, 6%). Variable nuclear accumulation of TP53, mirrored by elevated nuclear MDM2 expression was seen in >50% of cases. Furthermore, sinonasal melanomas (n = 7) including RAS/BRAF-wild type tumors (n = 5) harbored alterations of the key components and regulators of canonical WNT-pathway: APC (4/90, 4%), CTNNB1 (3/90, 3%) and AMER1 (1/90, 1%). Both, TERT promoter mutations (5/53, 9%) and fusions (2/40, 5%) were identified. The latter occurred in BRAF/RAS-wild type tumors. No oncogenic fusion gene transcripts previously reported in cutaneous melanomas were detected. Eight tumors including 7 BRAF/RAS-wild type cases expressed ADCK4::NUMBL cis-fusion transcripts. In summary, this study documented mutational activation of NRAS and other key components and regulators of Ras-MAPK signaling pathway such as SPRED1 in a majority of sinonasal melanomas.
Collapse
Affiliation(s)
- Małgorzata Chłopek
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.,Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.
| | | | | | - Alina Kuźniacka
- Department of Biology and Genetics, Medical University of Gdańsk, Gdańsk, Poland
| | - Kinga Hińcza
- Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Kamila Kubicka
- Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Maciej Kaczorowski
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.,Department of Clinical and Experimental Pathology, Wrocław Medical University, Wrocław, Poland
| | - Michael Newford
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Yalan Liu
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Abbas Agaimy
- Institute of Pathology, University Hospital of Erlangen, Erlangen, Germany
| | - Wojciech Biernat
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Monika Durzyńska
- Department of Pathology, The Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ireneusz Dziuba
- Faculty of Medicine, University of Technology, Katowice, Poland
| | - Arndt Hartmann
- Institute of Pathology, University Hospital of Erlangen, Erlangen, Germany
| | - Shingo Inaguma
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Japan
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Japan
| | - Janusz Kopczyński
- Department of Surgical Pathology, Holycross Cancer Center, Kielce, Poland
| | - Michal Michal
- Sikl's Department of Pathology, University Hospital, Charles University in Prague, Medical Faculty in Plzeň, Plzeň, Czech Republic
| | - Michael Michal
- Sikl's Department of Pathology, University Hospital, Charles University in Prague, Medical Faculty in Plzeň, Plzeň, Czech Republic
| | - Rafał Pęksa
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Monika Prochorec-Sobieszek
- Department of Pathology, The Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna Starzyńska
- Department of Oral Surgery, Medical University of Gdańsk, Gdańsk, Poland
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Japan
| | - Bartosz Wasąg
- Department of Biology and Genetics, Medical University of Gdańsk, Gdańsk, Poland
| | - Artur Kowalik
- Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland.,Division of Medical Biology, Institute of Biology Jan Kochanowski University, Kielce, Poland
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
118
|
Secretory carcinoma of the minor salivary gland in the lip with ETV6-NTRK3 fusion: case report and literature review. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY, MEDICINE, AND PATHOLOGY 2022. [DOI: 10.1016/j.ajoms.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
119
|
Nagel R, Pataskar A, Champagne J, Agami R. Boosting Antitumor Immunity with an Expanded Neoepitope Landscape. Cancer Res 2022; 82:3637-3649. [PMID: 35904353 PMCID: PMC9574376 DOI: 10.1158/0008-5472.can-22-1525] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 01/07/2023]
Abstract
Immune-checkpoint blockade therapy has been successfully applied to many cancers, particularly tumors that harbor a high mutational burden and consequently express a high abundance of neoantigens. However, novel approaches are needed to improve the efficacy of immunotherapy for treating tumors that lack a high load of classic genetically derived neoantigens. Recent discoveries of broad classes of nongenetically encoded and inducible neoepitopes open up new avenues for therapeutic development to enhance sensitivity to immunotherapies. In this review, we discuss recent work on neoantigen discovery, with an emphasis on novel classes of noncanonical neoepitopes.
Collapse
Affiliation(s)
- Remco Nagel
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Abhijeet Pataskar
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Julien Champagne
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Reuven Agami
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Erasmus MC, Rotterdam University, Rotterdam, the Netherlands
| |
Collapse
|
120
|
Therapeutic Advances of Rare ALK Fusions in Non-Small Cell Lung Cancer. Curr Oncol 2022; 29:7816-7831. [PMID: 36290895 PMCID: PMC9601201 DOI: 10.3390/curroncol29100618] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 01/11/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases and is the leading cause of cancer-related death. Despite advances in chemotherapy and immunotherapy, the prognosis for advanced patients remains poor. The discovery of oncogenic driver mutations, such as anaplastic lymphoma kinase (ALK) mutations, means that a subset of patients has opportunities for targeted therapy. With the improvement of genetic testing coverage, more and more ALK fusion subtypes and ALK partners have been discovered, and more than 90 rare ALK fusion subtypes have been found in NSCLC. However, unlike the common fusion, echinoderm microtubule-associated protein-like 4 (EML4)-ALK, some rare ALK fusions such as striatin (STRN)-ALK and huntingtin interacting protein 1 (HIP1)-ALK, etc., the large-scale clinical data related to its efficacy are still immature. The clinical application of ALK-tyrosine kinase inhibitors (ALK-TKIs) mainly depends on the positivity of the ALK gene, regardless of the molecular characteristics of the fusion partner. Recent clinical studies in the ALK-positive NSCLC population have demonstrated differences in progression-free survival (PFS) among patients based on different ALK fusion subtypes. This article will introduce the biological characteristics of ALK fusion kinase and common detection methods of ALK fusion and focus on summarizing the differential responses of several rare ALK fusions to ALK-TKIs, and propose corresponding treatment strategies, so as to better guide the application of ALK-TKIs in rare ALK fusion population.
Collapse
|
121
|
Moes-Sosnowska J, Skupinska M, Lechowicz U, Szczepulska-Wojcik E, Skronska P, Rozy A, Stepniewska A, Langfort R, Rudzinski P, Orlowski T, Popiel D, Stanczak A, Wieczorek M, Chorostowska-Wynimko J. FGFR1-4 RNA-Based Gene Alteration and Expression Analysis in Squamous Non-Small Cell Lung Cancer. Int J Mol Sci 2022; 23:ijms231810506. [PMID: 36142417 PMCID: PMC9505002 DOI: 10.3390/ijms231810506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/27/2022] [Accepted: 09/02/2022] [Indexed: 02/07/2023] Open
Abstract
While fibroblast growth factor receptors (FGFRs) are involved in several biological pathways and FGFR inhibitors may be useful in the treatment of squamous non-small cell lung cancer (Sq-NSCLC), FGFR aberrations are not well characterized in Sq-NSCLC. We comprehensively evaluated FGFR expression, fusions, and variants in 40 fresh-frozen primary Sq-NSCLC (stage IA3−IV) samples and tumor-adjacent normal tissues using real-time PCR and next-generation sequencing (NGS). Protein expression of FGFR1−3 and amplification of FGFR1 were also analyzed. FGFR1 and FGFR4 median gene expression was significantly (p < 0.001) decreased in tumors compared with normal tissue. Increased FGFR3 expression enhanced the recurrence risk (hazard ratio 4.72, p = 0.029), while high FGFR4 expression was associated with lymph node metastasis (p = 0.036). Enhanced FGFR1 gene expression was correlated with FGFR1 protein overexpression (r = 0.75, p = 0.0003), but not with FGFR1 amplification. NGS revealed known pathogenic FGFR2,3 variants, an FGFR3::TACC3 fusion, and a novel TACC1::FGFR1 fusion together with FGFR1,2 variants of uncertain significance not previously reported in Sq-NSCLC. These findings expand our knowledge of the Sq-NSCLC molecular background and show that combining different methods increases the rate of FGFR aberrations detection, which may improve patient selection for FGFRi treatment.
Collapse
MESH Headings
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Microtubule-Associated Proteins
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 4/genetics
Collapse
Affiliation(s)
- Joanna Moes-Sosnowska
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland or
| | - Monika Skupinska
- Preclinical Development Department, Celon Pharma S.A, Research & Development Centre, 05-152 Kazun Nowy, Poland
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland or
| | - Ewa Szczepulska-Wojcik
- Department of Pathology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| | - Paulina Skronska
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland or
| | - Adriana Rozy
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland or
| | - Aneta Stepniewska
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland or
| | - Renata Langfort
- Department of Pathology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| | - Piotr Rudzinski
- Department of Surgery, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| | - Tadeusz Orlowski
- Department of Surgery, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| | - Delfina Popiel
- Preclinical Development Department, Celon Pharma S.A, Research & Development Centre, 05-152 Kazun Nowy, Poland
| | - Aleksandra Stanczak
- Clinical Development Department, Celon Pharma S.A., Research & Development Centre, 05-152 Kazun Nowy, Poland
| | - Maciej Wieczorek
- Preclinical Development Department, Celon Pharma S.A, Research & Development Centre, 05-152 Kazun Nowy, Poland
- Clinical Development Department, Celon Pharma S.A., Research & Development Centre, 05-152 Kazun Nowy, Poland
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland or
- Correspondence: or
| |
Collapse
|
122
|
Ito T, Kinoshita K, Tomizawa M, Shinohara S, Nishii H, Matsushita M, Hattori K, Kohchi Y, Kohchi M, Hayase T, Watanabe F, Hasegawa K, Tanaka H, Kuramoto S, Takanashi K, Oikawa N. Discovery of CH7057288 as an Orally Bioavailable, Selective, and Potent pan-TRK Inhibitor. J Med Chem 2022; 65:12427-12444. [PMID: 36066182 DOI: 10.1021/acs.jmedchem.2c01099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Kinase fusions involving tropomyosin receptor kinases (TRKs) have been proven to act as strong oncogenic drivers and are therefore recognized as attractive therapeutic targets. We screened an in-house kinase-focused library and identified a promising hit compound with a unique tetracyclic scaffold. Compound 1 showed high TRK selectivity but moderate cell growth inhibitory activity as well as a potential risk of inducing CYP3A4. In this report, chemical modification intended to improve TRK inhibition and avoid CYP3A4 induction enabled us to identify an orally bioavailable, selective, and potent TRK inhibitor 7.
Collapse
Affiliation(s)
- Toshiya Ito
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Kazutomo Kinoshita
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Masaki Tomizawa
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Shojiro Shinohara
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Hiroki Nishii
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Masayuki Matsushita
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Kazuo Hattori
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Yasunori Kohchi
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Masami Kohchi
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Tadakatsu Hayase
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Fumio Watanabe
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Kiyoshi Hasegawa
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Hiroshi Tanaka
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Shino Kuramoto
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Kenji Takanashi
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Nobuhiro Oikawa
- Research Division, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| |
Collapse
|
123
|
Rooper LM, Thompson LDR, Gagan J, Hwang JSG, London NR, Mikula MW, Stevens TM, Bishop JA. Low-grade non-intestinal-type sinonasal adenocarcinoma: a histologically distinctive but molecularly heterogeneous entity. Mod Pathol 2022; 35:1160-1167. [PMID: 35322195 DOI: 10.1038/s41379-022-01068-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 11/10/2022]
Abstract
Although low-grade non-intestinal-type sinonasal adenocarcinoma (SNAC) is formally a diagnosis of exclusion defined by the absence of salivary or intestinal differentiation, most tumors in this category comprise a distinctive histologic group that are increasingly thought to derive from seromucinous glands. However, the molecular underpinnings of SNAC remain poorly understood, and it is unclear if diverse genetic alterations recently reported in isolated cases should delineate separate subgroups. This study aims to perform comprehensive evaluation of gene fusions and mutations and their histologic correlates in low-grade SNAC to clarify its pathogenesis and classification. We identified 18 non-intestinal-type SNAC that all displayed characteristic tubulopapillary architecture and low-grade cytology, although several cases had other unique histologic features and 3 showed intermixed high-grade areas. Among tumors stained with S100 protein, SOX10, and DOG1, 86% expressed at least one of these seromucinous markers. Of 17 cases with sufficient RNA or DNA available for analysis, likely oncogenic molecular alterations were identified in 76% of cases, most notably including CTNNB1 p.S33F mutations in 2 cases, concomitant BRAF p.V600E and AKT1 p.E17K mutations in 2 cases, and ETV6::NTRK3, PRKAR1A::MET, FN1::NRG1, and DNAJB1::PRKACA fusions in 1 case each. While tumors with most genetic alterations were histologically indistinguishable, cases with CTNNB1 mutations had intermixed squamoid morules and cases with BRAF and AKT1 mutations showed a myoepithelial cell population and prominent papillary to micropapillary architecture. Overall, these findings confirm previous reports of frequent seromucinous differentiation in low-grade SNAC. However, these tumors display striking molecular diversity with involvement of multiple kinase fusions, leading to frequent activation of signaling cascades including the MAPK pathway. While most genetic alterations are not associated with sufficiently distinctive histologic features to suggest separate classification, biphasic tumors with BRAF p.V600E mutations are more unique and may represent a distinctive subgroup.
Collapse
Affiliation(s)
- Lisa M Rooper
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Nyall R London
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael W Mikula
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Todd M Stevens
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
124
|
Velaga R, Koo KM, Mainwaring PN. Harnessing gene fusion-derived neoantigens for 'cold' breast and prostate tumor immunotherapy. Immunotherapy 2022; 14:1165-1179. [PMID: 36043380 DOI: 10.2217/imt-2022-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Breast and prostate cancers are generally considered immunologically 'cold' tumors due to multiple mechanisms rendering them unresponsive to immune checkpoint blockade therapies. With little success in garnering positive outcomes in modern immunotherapeutic clinical trials, it is prudent to re-examine the role of immunogenic neoantigens in these cold tumors. Gene fusions are driver mutations in hormone-driven cancers that can result in alternative mutation-specific neoantigens to promote immunotherapy sensitivity. This review focuses on 1) gene fusion formation mechanisms in neoantigen generation; 2) gene fusion neoantigens in cancer immunotherapeutic strategies and associated clinical trials; and 3) challenges and opportunities in computational and liquid biopsy technologies. This review is anticipated to initiate further research into gene fusion neoantigens of cold tumors for further experimental validation.
Collapse
Affiliation(s)
- Ravi Velaga
- Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Kevin M Koo
- XING Technologies Pty Ltd, Brisbane, QLD 4073, Australia.,The University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD 4029, Australia
| | | |
Collapse
|
125
|
Bridgewater J, Jiao X, Parimi M, Flach C, Stratford J, Kamburov A, Schmitz AA, Zong J, Reeves JA, Keating K, Bruno A, Fellous M, Pereira MB, Bazhenova L. Prognosis and oncogenomic profiling of patients with tropomyosin receptor kinase fusion cancer in the 100,000 genomes project. Cancer Treat Res Commun 2022; 33:100623. [PMID: 36041373 DOI: 10.1016/j.ctarc.2022.100623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are oncogenic drivers in various tumor types. Limited data exist on the overall survival (OS) of patients with tumors with NTRK gene fusions and on the co-occurrence of NTRK fusions with other oncogenic drivers. MATERIALS AND METHODS This retrospective study included patients enrolled in the Genomics England 100,000 Genomes Project who had linked clinical data from UK databases. Patients who had undergone tumor whole genome sequencing between March 2016 and July 2019 were included. Patients with and without NTRK fusions were matched. OS was analyzed along with oncogenic alterations in ALK, BRAF, EGFR, ERBB2, KRAS, and ROS1, and tumor mutation burden (TMB) and microsatellite instability (MSI). RESULTS Of 15,223 patients analyzed, 38 (0.25%) had NTRK gene fusions in 11 tumor types, the most common were breast cancer, colorectal cancer (CRC), and sarcoma. Median OS was not reached in both the NTRK gene fusion-positive and -negative groups (hazard ratio 1.47, 95% CI 0.39-5.57, P = 0.572). A KRAS mutation was identified in two (5%) patients with NTRK gene fusions, and both had hepatobiliary cancer. High TMB and MSI were both more common in patients with NTRK gene fusions, due to the CRC subset. While there was a higher risk of death in patients with NTRK gene fusions compared to those without, the difference was not statistically significant. CONCLUSION This study supports the hypothesis that NTRK gene fusions are primary oncogenic drivers and the co-occurrence of NTRK gene fusions with other oncogenic alterations is rare.
Collapse
Affiliation(s)
- John Bridgewater
- University College London Hospitals NHS Trust, London, United Kingdom; University College London Cancer Institute, London, United Kingdom.
| | - Xiaolong Jiao
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ, United States of America
| | | | - Clare Flach
- Real World Solutions, IQVIA, London, United Kingdom
| | | | | | | | - Jihong Zong
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ, United States of America
| | - John A Reeves
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ, United States of America
| | - Karen Keating
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ, United States of America
| | - Amanda Bruno
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ, United States of America
| | - Marc Fellous
- Bayer HealthCare Pharmaceuticals, Inc., Basel, Switzerland
| | | | - Lyudmila Bazhenova
- University of California San Diego Moores Cancer Center, San Diego, CA, United States of America
| |
Collapse
|
126
|
Loddo M, Hardisty KM, Llewelyn A, Haddow T, Thatcher R, Williams G. Utilisation of semiconductor sequencing for detection of actionable fusions in solid tumours. PLoS One 2022; 17:e0246778. [PMID: 35984852 PMCID: PMC9390944 DOI: 10.1371/journal.pone.0246778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 07/22/2022] [Indexed: 11/19/2022] Open
Abstract
Oncogenic fusions represent compelling druggable targets in solid tumours highlighted by the recent site agnostic FDA approval of larotrectinib for NTRK rearrangements. However screening for fusions in routinely processed tissue samples is constrained due to degradation of nucleic acid as a result of formalin fixation., To investigate the clinical utility of semiconductor sequencing optimised for detection of actionable fusion transcripts in formalin fixed samples, we have undertaken an analysis of test trending data generated by a clinically validated next generation sequencing platform designed to capture 867 of the most clinically relevant druggable driver-partner oncogenic fusions. Here we show across a real-life cohort of 1112 patients with solid tumours that actionable fusions occur at high frequency (7.4%) with linkage to a wide range of targeted therapy protocols including seven fusion-drug matches with FDA/EMA approval and/or NCCN/ESMO recommendations and 80 clinical trials. The more prevalent actionable fusions identified were independent of tumour type in keeping with signalling via evolutionary conserved RAS/RAF/MEK/ERK, PI3K/AKT/MTOR, PLCy/PKC and JAK/STAT pathways. Taken together our data indicates that semiconductor sequencing for detection of actionable fusions can be integrated into routine diagnostic pathology workflows enabling the identification of personalised treatment options that have potential to improve clinical cancer management across many tumour types.
Collapse
Affiliation(s)
- Marco Loddo
- Oncologica UK Ltd, Cambridge, United Kingdom
- * E-mail: (ML); (GW)
| | | | | | | | | | - Gareth Williams
- Oncologica UK Ltd, Cambridge, United Kingdom
- * E-mail: (ML); (GW)
| |
Collapse
|
127
|
Liu ZH, Zhu BW, Shi M, Qu YR, He XJ, Yuan HL, Ma J, Li W, Zhao DD, Liu ZC, Wang BM, Wang CY, Tao HQ, Ma TH. Profiling of gene fusion involving targetable genes in Chinese gastric cancer. World J Gastrointest Oncol 2022; 14:1528-1539. [PMID: 36160735 PMCID: PMC9412921 DOI: 10.4251/wjgo.v14.i8.1528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/14/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Approximately half of all new cases of gastric cancer (GC) and related deaths occur in China. More than 80% of patients with GC are diagnosed at an advanced stage, which results in poor prognosis. Although HER2-directed therapy and immune checkpoint inhibitors have been somewhat successful, new drugs are still needed for the treatment of GC. Notably, several gene fusion-targeted drugs have been approved by the United States Food and Drug Administration for solid tumors, including GC, such as larotrectinib for NTRK fusion-positive cancers and zenocutuzumab for NRG1 fusion-positive cancers. However, gene fusions involving targetable genes have not been well characterized in Chinese patients with GC.
AIM To identify the profile of fusions involving targetable genes in Chinese patients with GC using clinical specimens and determine the distribution of patients with gene fusion variants among the molecular subtypes of GC.
METHODS We retrospectively analyzed gene fusion events in tumor tissue samples from 954 Chinese patients with GC. Clinicopathological characteristics were obtained from their medical records. Genetic alterations, such as single nucleotide variants, indels, amplifications, and gene fusions, were identified using a targeted sequencing panel containing 825 genes. Fusions were validated by fluorescence in situ hybridization (FISH) using break-apart probes. The microsatellite instability (MSI) status was evaluated using MSIsensor from the targeted sequencing panel data. Tumor mutational burden (TMB) was calculated using the total number of nonsynonymous mutations divided by the total genomic targeted region. Chi-square analysis was used to determine the enrichment of gene fusions associated with the molecular subtypes of GC.
RESULTS We found that 1.68% (16/954) of patients harbored 20 fusion events involving targetable genes. RARA fusions (n = 5) were the most common, followed by FGFR2, BRAF, MET, FGFR3, RET, ALK, EGFR, NTRK2, and NRG1 fusions. Two of the RARA fusions, EML4-ALK (E6:E20) and EGFR-SEPTIN14 (E7:E10), have been identified in other tumors but not in GC. Surprisingly, 18 gene fusion events were previously not reported in any cancer types. Twelve of the eighteen novel gene fusions included complete exons encoding functional domains of targetable genes, such as the tyrosine kinase domain of receptor tyrosine kinases and the DNA- and ligand-binding domains of RARA. Consistent with the results of detection using the targeted sequencing fusion panel, the results of FISH (fluorescence in situ hybridization) confirmed the rearrangement of FGFR2 and BRAF in tumors from patients 04 and 09, respectively. Genetic analysis indicated that the fusion genes were significantly enriched in patients with ERBB2 amplification (P = 0.02); however, there were no significant differences between fusion-positive and fusion-negative patients in age, sex, MSI status, and TMB.
CONCLUSION We characterized the landscape of fusions involving targetable genes in a Chinese GC cohort and found that 1.68% of patients with GC harbor potential targetable gene fusions, which were enriched in patients with ERBB2 amplification. Gene fusion detection may provide a potential treatment strategy for patients with GC with disease progression following standard therapy.
Collapse
Affiliation(s)
- Zhen-Hua Liu
- Department of Medical Oncology, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Bo-Wen Zhu
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Min Shi
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Yu-Rong Qu
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Xun-Jun He
- Department of Genetics and Genomic Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Hong-Ling Yuan
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Jie Ma
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Wei Li
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Dan-Dan Zhao
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Zheng-Chuang Liu
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Bao-Ming Wang
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Chun-Yang Wang
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Hou-Quan Tao
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Tong-Hui Ma
- Department of Translational Medicine, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| |
Collapse
|
128
|
FGFR3-TACCs3 Fusions and Their Clinical Relevance in Human Glioblastoma. Int J Mol Sci 2022; 23:ijms23158675. [PMID: 35955806 PMCID: PMC9369421 DOI: 10.3390/ijms23158675] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023] Open
Abstract
Oncogenic fusion genes have emerged as successful targets in several malignancies, such as chronic myeloid leukemia and lung cancer. Fusion of the fibroblast growth receptor 3 and the transforming acidic coiled coil containing protein—FGFR3-TACC3 fusion—is prevalent in 3–4% of human glioblastoma. The fusion protein leads to the constitutively activated kinase signaling of FGFR3 and thereby promotes cell proliferation and tumor progression. The subgroup of FGFR3-TACC3 fusion-positive glioblastomas presents with recurrent clinical and histomolecular characteristics, defining a distinctive subtype of IDH-wildtype glioblastoma. This review aims to provide an overview of the available literature on FGFR3-TACC3 fusions in glioblastoma and possible implications for actual clinical practice.
Collapse
|
129
|
Experimentally Deduced Criteria for Detection of Clinically Relevant Fusion 3′ Oncogenes from FFPE Bulk RNA Sequencing Data. Biomedicines 2022; 10:biomedicines10081866. [PMID: 36009413 PMCID: PMC9405289 DOI: 10.3390/biomedicines10081866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/15/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Drugs targeting receptor tyrosine kinase (RTK) oncogenic fusion proteins demonstrate impressive anti-cancer activities. The fusion presence in the cancer is the respective drug prescription biomarker, but their identification is challenging as both the breakpoint and the exact fusion partners are unknown. RNAseq offers the advantage of finding both fusion parts by screening sequencing reads. Paraffin (FFPE) tissue blocks are the most common way of storing cancer biomaterials in biobanks. However, finding RTK fusions in FFPE samples is challenging as RNA fragments are short and their artifact ligation may appear in sequencing libraries. Here, we annotated RNAseq reads of 764 experimental FFPE solid cancer samples, 96 leukemia samples, and 2 cell lines, and identified 36 putative clinically relevant RTK fusions with junctions corresponding to exon borders of the fusion partners. Where possible, putative fusions were validated by RT-PCR (confirmed for 10/25 fusions tested). For the confirmed 3′RTK fusions, we observed the following distinguishing features. Both moieties were in-frame, and the tyrosine kinase domain was preserved. RTK exon coverage by RNAseq reads upstream of the junction site were lower than downstream. Finally, most of the true fusions were present by more than one RNAseq read. This provides the basis for automatic annotation of 3′RTK fusions using FFPE RNAseq profiles.
Collapse
|
130
|
Myer NM, Shitara K, Chung HC, Lordick F, Kelly RJ, Szabo Z, Cao ZA, Leong S, Ilson DH, Weichert W. Evolution of predictive and prognostic biomarkers in the treatment of advanced gastric cancer. J Cancer Res Clin Oncol 2022; 148:2023-2043. [PMID: 35551464 PMCID: PMC11110882 DOI: 10.1007/s00432-021-03902-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022]
Abstract
Despite new therapeutic options, advanced gastric cancer remains associated with a poor prognosis compared with other cancers. Recent gains in the treatment of gastric cancer were accompanied by the identification of novel biomarkers associated with various cellular pathways and corresponding diagnostic technologies. It is expected that the standardization of clinical workflow and technological refinements in biomarker assessment will support greater personalization and further improve treatment outcomes. In this article, we review the current state of prognostic and predictive biomarkers in gastric cancer.
Collapse
Affiliation(s)
- Nicole M Myer
- Merck & Co., Inc., 90 E. Scott Avenue, Rahway, NJ, 07065, USA.
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Hyun C Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Florian Lordick
- Medical Department (Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases), University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ronan J Kelly
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Zsolt Szabo
- Merck & Co., Inc., Ringstrasse 27 Kriens, LUZERN, 6010, Switzerland
| | - Z Alexander Cao
- Merck & Co., Inc., 90 E. Scott Avenue, Rahway, NJ, 07065, USA
| | - Stephen Leong
- Merck & Co., Inc., 351 N Sumneytown Pike, North Wales, PA, 19454, USA
| | - David H Ilson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wilko Weichert
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
131
|
Gros L, Dei Tos AP, Jones RL, Digklia A. Inflammatory Myofibroblastic Tumour: State of the Art. Cancers (Basel) 2022; 14:cancers14153662. [PMID: 35954326 PMCID: PMC9367282 DOI: 10.3390/cancers14153662] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Among sarcomas, which are rare cancers, inflammatory myofibroblastic tumors are extremely rare. Unlike other subtypes, this is a largely oncogene-driven neoplasia, and early gene rearrangement identification is important for accurate advanced stage treatment. In this manuscript, we review the clinicopathologic characteristics of this ultra-rare entity, as well as the current treatment landscape, with a particular focus on opportunities provided by tyrosine kinase inhibitors (TKIs). Abstract An inflammatory myofibroblastic tumor (IMT) is a neoplasm composed of myofibroblastic and fibroblastic spindle cells accompanied by inflammatory cells, including lymphocytes and eosinophils. It is an ultra-rare tumor, the optimal management of which remains to be defined. Surgery is the treatment of choice for localized tumors. The treatment of advanced disease is not precisely defined. Chemotherapy regimens result in an overall response rate of approximately 50% based on retrospective data. The latest pathophysiological data highlight the role played by tyrosine kinase fusion genes in IMT proliferation. Anaplast lymphoma kinase (ALK) oncogenic activation mechanisms have been characterized in approximately 80% of IMTs. In this context, data regarding targeted therapies are most important. The aims of this article are to review the latest published data on the use of systematic therapy, particularly the use of molecular targeted therapy, and to publish an additional case of an IMT with Ran-binding protein 2 (RANPB2)-ALK fusion showing a long response to a tyrosine kinase inhibitor.
Collapse
Affiliation(s)
- Louis Gros
- Department of Oncology, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| | - Angelo Paolo Dei Tos
- Department of Pathology, Azienda Ospedale Università Padova, 35128 Padua, Italy;
- Department of Medicine, University of Padua School of Medicine, 35128 Padua, Italy
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK;
- Division of Clinical Sciences, Institute of Cancer Research, Royal Marsden Hospital, London SW3 6JJ, UK
| | - Antonia Digklia
- Department of Oncology, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
- Center of Sarcoma, Department of Oncology, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
- Correspondence:
| |
Collapse
|
132
|
Imai M, Nakamura Y, Sunami K, Kage H, Komine K, Koyama T, Amano T, Ennishi D, Kanai M, Kenmotsu H, Maeda T, Morita S, Sakai D, Bando H, Makiyama A, Suzuki T, Hirata M, Kohsaka S, Tsuchihara K, Naito Y, Yoshino T. Expert Panel Consensus Recommendations on the Use of Circulating Tumor DNA Assays for Patients with Advanced Solid Tumors. Cancer Sci 2022; 113:3646-3656. [PMID: 35876224 PMCID: PMC9633310 DOI: 10.1111/cas.15504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 12/01/2022] Open
Abstract
Comprehensive genomic profiling is increasingly used to facilitate precision oncology based on molecular stratification. In addition to conventional tissue comprehensive genomic profiling, comprehensive genomic profiling of circulating tumor DNA has become widely utilized in cancer care owing on its advantages, including less invasiveness, rapid turnaround time, and capturing heterogeneity. However, circulating tumor DNA comprehensive genomic profiling has some limitations, mainly false negatives due to low levels of plasma circulating tumor deoxyribonucleic acid and false positives caused by clonal hematopoiesis. Nevertheless, no guidelines and recommendations fully address these issues. Here, an expert panel committee involving representatives from 12 Designated Core Hospitals for Cancer Genomic Medicine in Japan was organized to develop expert consensus recommendations for the use of circulating tumor deoxyribonucleic acid‐based comprehensive genomic profiling. The aim was to generate guidelines for clinicians and allied healthcare professionals on the optimal use of the circulating tumor DNA assays in advanced solid tumors and to aid the design of future clinical trials that utilize and develop circulating tumor DNA assays to refine precision oncology. Fourteen clinical questions regarding circulating tumor deoxyribonucleic acid comprehensive genomic profiling including the timing of testing and considerations for interpreting results were established by searching and curating associated literatures, and corresponding recommendations were prepared based on the literature for each clinical question. Final consensus recommendations were developed by voting to determine the level of each recommendation by the Committee members.
Collapse
Affiliation(s)
- Mitsuho Imai
- Translational Research Support Section, National Cancer Center Hospital East.,Genomics Unit, Keio University School of Medicine
| | - Yoshiaki Nakamura
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital
| | - Hidenori Kage
- Department of Next-Generation Precision Medicine Development Laboratory, Graduate School of Medicine, The University of Tokyo
| | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital
| | - Toraji Amano
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University
| | | | - Takahiro Maeda
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences
| | - Sachi Morita
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital
| | - Daisuke Sakai
- Center for Cancer Genomics and Personalized Medicine, Osaka University Hospital
| | - Hideaki Bando
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| | | | - Tatsuya Suzuki
- Department of Hematology, National Cancer Center Hospital
| | - Makoto Hirata
- Department of Genetic Medicine and Services, National Cancer Center Hospital
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Japan
| | - Yoichi Naito
- Department of General Internal medicine/Experimental Therapeutics/Medical Oncology, National Cancer Center Hospital East
| | - Takayuki Yoshino
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| |
Collapse
|
133
|
Wang Z, Ren J, Jia K, Zhao Y, Liang L, Cheng Z, Huang F, Zhao X, Cheng J, Song S, Sheng T, Wan W, Shu Q, Wu D, Zhang J, Lu T, Chen Y, Ran T, Lu S. Identification and structural analysis of a selective tropomyosin receptor kinase C (TRKC) inhibitor. Eur J Med Chem 2022; 241:114601. [PMID: 35872544 DOI: 10.1016/j.ejmech.2022.114601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 07/07/2022] [Indexed: 11/04/2022]
Abstract
Tropomyosin receptor kinases (TRKs) are a family of TRKA, TRKB and TRKC isoforms. It has been widely reported that TRKs are implicated in a variety of tumors with several Pan-TRK inhibitors currently being used or evaluated in clinical treatment. However, off-target adverse events frequently occur in the clinical use of Pan-TRK inhibitors, which result in poor patient compliance, even drug discontinuation. Although a subtype-selectivity TRK inhibitor may avert the potential off-target adverse events and can act as a more powerful tool compound in the biochemical studies on TRKs, the high sequence similarities of TRKs hinder the development of subtype-selectivity TRK inhibitors. For example, no selective TRKC inhibitor has been reported. Herein, a selective TRKC inhibitor (L13) was disclosed, with potent TRKC inhibitory activity and 107.5-/34.9-fold selectivity over TRKA/B (IC50 TRKA/B/C = 1400 nM, 454 nM, 13 nM, respectively). Extensive molecular dynamics simulations illustrated that key interactions of L13 with the residues and diversely conserved water molecules in the ribose regions of different TRKs may be the structural basis of selectivity. This will provide inspiring insights into the development of subtype-selectivity TRK inhibitors. Moreover, L13 could serve as a tool compound to investigate the distinct biological functions of TRKC and a starting point for further research on drugs specifically targeting TRKC.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jiwei Ren
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Kun Jia
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yuming Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, PR China
| | - Li Liang
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Zitian Cheng
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Fei Huang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Xiaofei Zhao
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Shiyu Song
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, 210038, PR China
| | - Tiancheng Sheng
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Weiqi Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Qingqing Shu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Donglin Wu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Junhao Zhang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Ting Ran
- Drug and Vaccine Research Center, Guangzhou Laboratory, Guangzhou, 510005, PR China.
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
134
|
Oncogenic Signalling of Growth Factor Receptors in Cancer: Mechanisms and Therapeutic Opportunities. Int J Mol Sci 2022; 23:ijms23137376. [PMID: 35806381 PMCID: PMC9266644 DOI: 10.3390/ijms23137376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/29/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer is a common name for several distinct diseases caused by uncontrolled cell growth and proliferation [...]
Collapse
|
135
|
Wang L, Zhang D, Luo Z, Feng J, Liao W, Li J, Wang J. Gram-scale stereoselective synthesis of next generation of Trk Inhibitor LOXO-195. Tetrahedron Lett 2022. [DOI: 10.1016/j.tetlet.2022.154019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
136
|
Manea CA, Badiu DC, Ploscaru IC, Zgura A, Bacinschi X, Smarandache CG, Serban D, Popescu CG, Grigorean VT, Botnarciuc V. A review of NTRK fusions in cancer. Ann Med Surg (Lond) 2022; 79:103893. [PMID: 35860155 PMCID: PMC9289232 DOI: 10.1016/j.amsu.2022.103893] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/25/2022] [Accepted: 05/29/2022] [Indexed: 11/25/2022] Open
|
137
|
Kimura S, Montefiori L, Iacobucci I, Zhao Y, Gao Q, Paietta EM, Haferlach C, Laird AD, Mead PE, Gu Z, Stock W, Litzow M, Rowe JM, Luger SM, Hunger SP, Ryland GL, Schmidt B, Ekert PG, Oshlack A, Grimmond SM, Rehn J, Breen J, Yeung D, White DL, Aldoss I, Jabbour EJ, Pui CH, Meggendorfer M, Walter W, Kern W, Haferlach T, Brady S, Zhang J, Roberts KG, Blombery P, Mullighan CG. Enhancer retargeting of CDX2 and UBTF::ATXN7L3 define a subtype of high-risk B-progenitor acute lymphoblastic leukemia. Blood 2022; 139:3519-3531. [PMID: 35192684 PMCID: PMC9203703 DOI: 10.1182/blood.2022015444] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/12/2022] [Indexed: 01/14/2023] Open
Abstract
Transcriptome sequencing has identified multiple subtypes of B-progenitor acute lymphoblastic leukemia (B-ALL) of prognostic significance, but a minority of cases lack a known genetic driver. Here, we used integrated whole-genome (WGS) and -transcriptome sequencing (RNA-seq), enhancer mapping, and chromatin topology analysis to identify previously unrecognized genomic drivers in B-ALL. Newly diagnosed (n = 3221) and relapsed (n = 177) B-ALL cases with tumor RNA-seq were studied. WGS was performed to detect mutations, structural variants, and copy number alterations. Integrated analysis of histone 3 lysine 27 acetylation and chromatin looping was performed using HiChIP. We identified a subset of 17 newly diagnosed and 5 relapsed B-ALL cases with a distinct gene expression profile and 2 universal and unique genomic alterations resulting from aberrant recombination-activating gene activation: a focal deletion downstream of PAN3 at 13q12.2 resulting in CDX2 deregulation by the PAN3 enhancer and a focal deletion of exons 18-21 of UBTF at 17q21.31 resulting in a chimeric fusion, UBTF::ATXN7L3. A subset of cases also had rearrangement and increased expression of the PAX5 gene, which is otherwise uncommon in B-ALL. Patients were more commonly female and young adult with median age 35 (range,12-70 years). The immunophenotype was characterized by CD10 negativity and immunoglobulin M positivity. Among 16 patients with known clinical response, 9 (56.3%) had high-risk features including relapse (n = 4) or minimal residual disease >1% at the end of remission induction (n = 5). CDX2-deregulated, UBTF::ATXN7L3 rearranged (CDX2/UBTF) B-ALL is a high-risk subtype of leukemia in young adults for which novel therapeutic approaches are required.
Collapse
Affiliation(s)
- Shunsuke Kimura
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Lindsey Montefiori
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Yaqi Zhao
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Qingsong Gao
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | | | | | | | - Paul E Mead
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Zhaohui Gu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
- Departments of Computational and Quantitative Medicine, and Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA
| | - Wendy Stock
- Department of Hematology and Oncology, University of Chicago, Chicago, IL
| | - Mark Litzow
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Jacob M Rowe
- Department of Hematology, Rambam Health Care Campus, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Selina M Luger
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Stephen P Hunger
- Department of Pediatrics, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Georgina L Ryland
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Precision Oncology, Centre for Cancer Research, and
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Breon Schmidt
- Computational Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Paul G Ekert
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre and School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Alicia Oshlack
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Parkville, VIC, Australia
| | | | - Jacqueline Rehn
- Blood Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - James Breen
- Blood Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - David Yeung
- Blood Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Hematology Department, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Deborah L White
- Blood Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA
| | - Elias J Jabbour
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | - Samuel Brady
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN
| | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Piers Blombery
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia; and
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
- Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
138
|
Dai Y, Liu P, He W, Yang L, Ni Y, Ma X, Du F, Song C, Liu Y, Sun Y. Genomic Features of Solid Tumor Patients Harboring ALK/ROS1/NTRK Gene Fusions. Front Oncol 2022; 12:813158. [PMID: 35785159 PMCID: PMC9243239 DOI: 10.3389/fonc.2022.813158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fusions of receptor tyrosine kinase (RTK) involving anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1), and neurotrophic receptor tyrosine kinase (NTRK) represent the potential targets of therapeutic intervention for various types of solid tumors. Here, the genomic features of 180 Chinese solid tumor patients with ALK, ROS1, and NTRK fusions by next generation sequencing (NGS) were comprehensively characterized, and the data from 121 patients in Memorial Sloan Kettering Cancer Center (MSKCC) database were used to compare. We found that ALK, ROS1, and NTRK fusions were more common in younger female patients (p<0.001) and showed a higher expression of programmed death ligand 1 (PD-L1). The gene-intergenic fusion and the fusion with rare formation directions accounted for a certain proportion in all samples and 62 novel fusions were discovered. Alterations in TP53 and MUC16 were common in patients with RTK fusions. The mutational signatures of patients were mainly distributed in COSMIC signature 1, 2, 3, 15 and 30, while had a higher frequency in copy number variations (CNVs) of individual genes, such as IL-7R. In the MSKCC cohort, patients with fusions and CNVs showed shorter overall survival than those with only fusions. Furthermore, the differentially mutated genes between fusion-positive and -negative patients mainly concentrated on MAPK signaling and FOXO signaling pathways. These results may provide genomic information for the personalized clinical management of solid tumor patients with ALK, ROS1, and NTRK fusions in the era of precision medicine.
Collapse
Affiliation(s)
- Yinghuan Dai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ping Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenlong He
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lizhen Yang
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yang Ni
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Xuejiao Ma
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Furong Du
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Chao Song
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
- *Correspondence: Yi Sun, ; Yang Liu, ; Chao Song,
| | - Yang Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Yi Sun, ; Yang Liu, ; Chao Song,
| | - Yi Sun
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yi Sun, ; Yang Liu, ; Chao Song,
| |
Collapse
|
139
|
Gounder MM, Agaram NP, Trabucco SE, Robinson V, Ferraro RA, Millis SZ, Krishnan A, Lee J, Attia S, Abida W, Drilon A, Chi P, Angelo SPD, Dickson MA, Keohan ML, Kelly CM, Agulnik M, Chawla SP, Choy E, Chugh R, Meyer CF, Myer PA, Moore JL, Okimoto RA, Pollock RE, Ravi V, Singh AS, Somaiah N, Wagner AJ, Healey JH, Frampton GM, Venstrom JM, Ross JS, Ladanyi M, Singer S, Brennan MF, Schwartz GK, Lazar AJ, Thomas DM, Maki RG, Tap WD, Ali SM, Jin DX. Clinical genomic profiling in the management of patients with soft tissue and bone sarcoma. Nat Commun 2022; 13:3406. [PMID: 35705558 PMCID: PMC9200814 DOI: 10.1038/s41467-022-30496-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023] Open
Abstract
There are more than 70 distinct sarcomas, and this diversity complicates the development of precision-based therapeutics for these cancers. Prospective comprehensive genomic profiling could overcome this challenge by providing insight into sarcomas' molecular drivers. Through targeted panel sequencing of 7494 sarcomas representing 44 histologies, we identify highly recurrent and type-specific alterations that aid in diagnosis and treatment decisions. Sequencing could lead to refinement or reassignment of 10.5% of diagnoses. Nearly one-third of patients (31.7%) harbor potentially actionable alterations, including a significant proportion (2.6%) with kinase gene rearrangements; 3.9% have a tumor mutational burden ≥10 mut/Mb. We describe low frequencies of microsatellite instability (<0.3%) and a high degree of genome-wide loss of heterozygosity (15%) across sarcomas, which are not readily explained by homologous recombination deficiency (observed in 2.5% of cases). In a clinically annotated subset of 118 patients, we validate actionable genetic events as therapeutic targets. Collectively, our findings reveal the genetic landscape of human sarcomas, which may inform future development of therapeutics and improve clinical outcomes for patients with these rare cancers.
Collapse
Affiliation(s)
- Mrinal M Gounder
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | | | | | | | - Richard A Ferraro
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | - Anita Krishnan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Lee
- Foundation Medicine, Inc., Cambridge, MA, USA
| | | | - Wassim Abida
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ping Chi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Sandra P D' Angelo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Mark A Dickson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Mary Lou Keohan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ciara M Kelly
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | - Sant P Chawla
- Sarcoma Center of Santa Monica, Santa Monica, CA, USA
| | - Edwin Choy
- Massachusetts General Hospital, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Christian F Meyer
- Johns Hopkins Sidney Kimmel Comprehensive Center, Baltimore, MD, USA
| | - Parvathi A Myer
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Ross A Okimoto
- University of California at San Francisco, San Francisco, CA, USA
| | | | - Vinod Ravi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arun S Singh
- University of California at Los Angeles, Los Angeles, CA, USA
| | - Neeta Somaiah
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Wagner
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - John H Healey
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | | | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, MA, USA
- Albany Medical College, Albany, NY, USA
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel Singer
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Murray F Brennan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Gary K Schwartz
- Herbert Irving Cancer Center, Columbia University, New York, NY, USA
| | | | - David M Thomas
- Garvan Institute of Medical Research, Darlinghurst,, NSW, Australia
| | - Robert G Maki
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - William D Tap
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, MA, USA
| | | |
Collapse
|
140
|
Yang JCH, Brose MS, Castro G, Kim ES, Lassen UN, Leyvraz S, Pappo A, López-Ríos F, Reeves JA, Fellous M, Penault-Llorca F, Rudzinski ER, Tabatabai G, Vassal G, Drilon A, Trent J. Rationale and design of ON-TRK: a novel prospective non-interventional study in patients with TRK fusion cancer treated with larotrectinib. BMC Cancer 2022; 22:625. [PMID: 35672677 PMCID: PMC9171956 DOI: 10.1186/s12885-022-09687-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tropomyosin receptor kinase (TRK) fusion proteins resulting from neurotrophic tyrosine receptor kinase (NTRK) gene fusions are rare primary oncogenic drivers in a wide array of tumors. Larotrectinib is a first-in-class, highly selective, central nervous system-active TRK inhibitor approved by the US Food and Drug Administration (FDA), European Medicines Agency (EMA), and over 40 countries for the treatment of TRK fusion solid tumors in adult and pediatric patients. Due to the rarity of TRK fusion cancer, larotrectinib was granted accelerated approval based on a relatively small number of patients enrolled in three early phase trials. ON-TRK aims to evaluate the safety profile of larotrectinib in a broader population and over extended time periods. METHODS ON-TRK is a prospective, non-interventional, open-label, multicenter, multi-cohort, post-approval study in adult and pediatric patients with locally advanced or metastatic TRK fusion cancer treated with larotrectinib that will describe the safety and effectiveness of larotrectinib in real-world practice conditions. Adult patients will be grouped by tumor type and followed for at least 2 years. Patients < 18 years old will be enrolled under a 'pediatric' cohort regardless of tumor type and will be followed for 5 years to evaluate the risk of potential long-term adverse effects of larotrectinib on their growth and development. The effectiveness of larotrectinib in the overall study population as well as in patient subgroups will also be evaluated. Procedures avoided in patients with infantile fibrosarcoma (e.g., amputation) and the number of patients who were able to undergo surgery with a curative intent (excluding amputation) because of the use of larotrectinib will be described. Larotrectinib treatment patterns in real-world practice, including dosing and duration of treatment, will be described. DISCUSSION The FDA Accelerated Approval Program allows for earlier approval of and patient access to drugs that treat serious conditions and fill an unmet medical need. This study is designed to fulfill post-approval requirements set by the FDA as well as post-marketing requirements set forth by local regulatory bodies and is part of the risk management plan for the EMA. STUDY REGISTRATION This study is registered at ClinicalTrials.gov ( NCT04142437 ). PROTOCOL VERSION v2.5, 25 March 2021.
Collapse
Affiliation(s)
- James C H Yang
- National Taiwan University Cancer Center, Taipei City, Taiwan.
| | - Marcia S Brose
- Abramson Cancer Center of the University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Current affiliation: Sidney Kimmel Cancer Center of Jefferson University Health, Philadelphia, PA, USA
| | - Gilberto Castro
- Instituto Do Câncer Do Estado de São Paulo, São Paulo, Brazil
| | - Edward S Kim
- Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
- Current affiliation: City of Hope National Medical Center, Los Angeles, CA, USA
| | - Ulrik N Lassen
- Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Serge Leyvraz
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alberto Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Fernando López-Ríos
- Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, Madrid, Spain
- Current affiliation: Department of Pathology, "12 de Octubre" University Hospital, Madrid, Spain
| | - John A Reeves
- Bayer HealthCare Pharmaceuticals Inc., Whippany, NJ, USA
| | - Marc Fellous
- Bayer HealthCare Pharmaceuticals, Inc., Basel, Switzerland
| | - Frédérique Penault-Llorca
- Department of Pathology, Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Center Jean Perrin, Montalembert, Clermont-Ferrand, France
| | - Erin R Rudzinski
- Seattle Children's Hospital and University of Washington Medical Center, Seattle, WA, USA
| | - Ghazaleh Tabatabai
- Department of Neurology & Interdisciplinary Neuro-Oncology, University Hospital Tübingen, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | | | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jonathan Trent
- Sylvester Comprehensive Cancer Center at University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
141
|
Wang ZZ, Wang MS, Wang F, Shi XX, Huang W, Hao GF, Yang GF. Exploring the kinase-inhibitor fragment interaction space facilitates the discovery of kinase inhibitor overcoming resistance by mutations. Brief Bioinform 2022; 23:6596988. [PMID: 35649390 DOI: 10.1093/bib/bbac203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/07/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Protein kinases play crucial roles in many cellular signaling processes, making them become important targets for drug discovery. But drug resistance mediated by mutation puts a barrier to the therapeutic effect of kinase inhibitors. Fragment-based drug discovery has been successfully applied to overcome such resistance. However, the complicate kinase-inhibitor fragment interaction and fragment-to-lead process seriously limit the efficiency of kinase inhibitor discovery against resistance caused by mutation. Here, we constructed a comprehensive web platform KinaFrag for the fragment-based kinase inhibitor discovery to overcome resistance. The kinase-inhibitor fragment space was investigated from 7783 crystal kinase-inhibitor fragment complexes, and the structural requirements of kinase subpockets were analyzed. The core fragment-based virtual screening workflow towards specific subpockets was developed to generate new kinase inhibitors. A series of tropomyosin receptor kinase (TRK) inhibitors were designed, and the most potent compound YT9 exhibits up to 70-fold activity improvement than marketed drugs larotrectinib and selitrectinib against G595R, G667C and F589L mutations of TRKA. YT9 shows promising antiproliferative against tumor cells in vitro and effectively inhibits tumor growth in vivo for wild type TRK and TRK mutants. Our results illustrate the great potential of KinaFrag in the kinase inhibitor discovery to combat resistance mediated by mutation. KinaFrag is freely available at http://chemyang.ccnu.edu.cn/ccb/database/KinaFrag/.
Collapse
Affiliation(s)
- Zhi-Zheng Wang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| | - Ming-Shu Wang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| | - Fan Wang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| | - Xing-Xing Shi
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| | - Wei Huang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| | - Ge-Fei Hao
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, P. R. China
| |
Collapse
|
142
|
Abughanimeh O, Kaur A, El Osta B, Ganti AK. Novel targeted therapies for advanced non-small lung cancer. Semin Oncol 2022; 49:326-336. [PMID: 35414419 DOI: 10.1053/j.seminoncol.2022.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/05/2021] [Accepted: 03/12/2022] [Indexed: 11/11/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer accounting for almost 80%-85% of all lung cancer cases. Unfortunately, more than half of the patients will be diagnosed with advanced disease at the time of presentation, which makes their disease incurable. Historically, the 5 year overall survival for advanced NSCLC was 5%. However, there has been a significant increase in our understanding of the genetic basis of NSCLC, which has led to development of both immunotherapy and targeted therapy agents. This has improved the 5 year overall survival to become within the range of 15%-50% depending on certain mutations and biomarkers. Over the last decade the United States Food and Drug Administration (FDA) has approved almost 20 new targeted therapies and clinical trials are still undergoing to evaluate more novel agents. In this review, we will present recent updates on novel targeted therapies.
Collapse
Affiliation(s)
- Omar Abughanimeh
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center- Fred and Pamela Buffett Cancer Center, Omaha, NE
| | - Anahat Kaur
- Albert Einstein College of Medicine/Jacobi Medical Center, Bronx, NY
| | - Badi El Osta
- Department of Hematology and Oncology, Atlanta VA Health Care System, Decatur, GA; Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Apar Kishor Ganti
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center- Fred and Pamela Buffett Cancer Center, Omaha, NE; Division of Oncology and Hematology, VA Nebraska-Western Iowa Health Care System, Omaha, NE.
| |
Collapse
|
143
|
Hernandez-Prera JC, Saeed-Vafa D, Heidarian A, Gewandter K, Otto K, Wenig BM. Sclerosing Polycystic Adenoma: Conclusive Clinical and Molecular Evidence of Its Neoplastic Nature. Head Neck Pathol 2022; 16:416-426. [PMID: 34410594 PMCID: PMC9187789 DOI: 10.1007/s12105-021-01374-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/14/2021] [Indexed: 02/06/2023]
Abstract
Sclerosing polycystic adenosis, initially considered a non-neoplastic salivary gland lesion and classified as such in the 2017 WHO Classification of Head and Neck Tumors, has been the subject of controversy regarding its possible neoplastic nature. The reporting of recurrent PI3K pathway alteration represents evidence to support these lesions as being neoplastic and more appropriately referred to as sclerosing polycystic adenoma (SPA). Herein, we provide additional evidence that supports the classification of SPA as a true neoplasm. Eight cases of SPA were identified in our database and consultation files. All cases were subjected to PTEN immunohistochemistry (IHC) and next-generation sequencing (NGS). In addition, one patient underwent genetic counseling and germline testing. The cases included 5 men and 3 women with a mean age of 41 years (range 11-78) and all tumors arose in the parotid gland. One patient had multiple recurrences over a period of 2 years. Morphologically the tumors were circumscribed and characterized by an admixture of acini, ducts and cysts embedded in a fibrotic/sclerotic stroma. The cells lining the ducts and cysts showed variable granular, vacuolated, foamy and apocrine cytoplasmic features, as well as acinar cells contained intracytoplasmic brightly eosinophilic granules. The apocrine intraductal proliferations showed mild to moderate atypia in 6 cases. One case showed overt malignant morphology that ranged from intraductal carcinoma to invasive salivary duct carcinoma. Seven cases tested for PTEN IHC showed loss of nuclear expression in the acinar and ductal cells with retained PTEN expression in the myoepithelial cell and stroma. NGS detected PIK3CA or PIK3R1 genetic alterations in 7 cases, including a novel TFG-PIK3CA fusion. Coexisting PTEN mutations were seen in 4 cases, including in a patient with clinical stigmata of Cowden syndrome and confirmed by germline genetic testing. Our findings herein documented including recurrence of tumor, malignant transformation, high prevalence of PI3K pathway oncogenic alterations and the possible heretofore undescribed association with Cowden syndrome add support to classifying SPA as true neoplasms justifying their designation as adenoma, rather than adenosis.
Collapse
Affiliation(s)
- Juan C Hernandez-Prera
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | - Daryoush Saeed-Vafa
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Amin Heidarian
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Kristen Otto
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Bruce M Wenig
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| |
Collapse
|
144
|
Patient with multiple genetically distinct thyroid nodules including papillary thyroid carcinoma harboring novel YWHAG-BRAF fusion. Cancer Genet 2022; 266-267:51-56. [DOI: 10.1016/j.cancergen.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/07/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022]
|
145
|
Mingione VR, Foda Z, Paung Y, Philipose H, Rangwala AM, Shan Y, Seeliger MA. Validation of an allosteric binding site of Src kinase identified by unbiased ligand binding simulations. J Mol Biol 2022; 434:167628. [PMID: 35595169 DOI: 10.1016/j.jmb.2022.167628] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/13/2022] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
Abstract
Allostery plays a primary role in regulating protein activity, making it an important mechanism in human disease and drug discovery. Identifying allosteric regulatory sites to explore their biological significance and therapeutic potential is invaluable to drug discovery; however, identification remains a challenge. Allosteric sites are often "cryptic" without clear geometric or chemical features. Since allosteric regulatory sites are often less conserved in protein kinases than the orthosteric ATP binding site, allosteric ligands are commonly more specific than ATP competitive inhibitors. We present a generalizable computational protocol to predict allosteric ligand binding sites based on unbiased ligand binding simulation trajectories. We demonstrate the feasibility of this protocol by revisiting our previously published ligand binding simulations using the first identified viral proto-oncogene, Src kinase, as a model system. The binding paths for kinase inhibitor PP1 uncovered three metastable intermediate states before binding the high-affinity ATP-binding pocket, revealing two previously known allosteric sites and one novel site. Herein, we validate the novel site using a combination of virtual screening and experimental assays to identify a v-type allosteric small-molecule inhibitor that targets this novel site with specificity for Src over closely related kinases. This study provides a proof-of-concept for employing unbiased ligand binding simulations to identify cryptic allosteric binding sites and is widely applicable to other protein-ligand systems.
Collapse
Affiliation(s)
- Victoria R Mingione
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Zachariah Foda
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - YiTing Paung
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Hannah Philipose
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Aziz M Rangwala
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yibing Shan
- Antidote Health Foundation for Cure of Cancer, Cambridge, MA 02139, USA.
| | - Markus A Seeliger
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
146
|
Le X, Baik C, Bauman J, Gilbert J, Brose MS, Grilley-Olson JE, Patil T, McDermott R, Raez LE, Johnson JM, Shen L, Tahara M, Ho AL, Norenberg R, Dima L, Brega N, Drilon A, Hong DS. Larotrectinib Treatment for Patients With TRK Fusion-Positive Salivary Gland Cancers. Oncologist 2022; 29:oyac080. [PMID: 35536733 PMCID: PMC11144979 DOI: 10.1093/oncolo/oyac080] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/09/2022] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Larotrectinib is a first-in-class, highly selective, and central nervous system-active tropomyosin receptor kinase (TRK) inhibitor approved for the treatment of adult and pediatric patients with TRK fusion cancer. We report the efficacy and safety of larotrectinib in patients with TRK fusion-positive salivary gland cancers. PATIENTS AND METHODS Patients with TRK fusion-positive salivary gland cancer treated with larotrectinib were identified from two clinical trials (NCT02122913 and NCT02576431). Patients received larotrectinib 100 mg twice daily (BID) except for one patient who received 150 mg BID in the phase I trial. The primary endpoint was objective response rate (ORR) as assessed by the investigator using Response Evaluation Criteria in Solid Tumors version 1.1. RESULTS At the data cut-off (July 20, 2020), 24 patients with TRK fusion-positive salivary gland cancer had been treated. The most common histologies were secretory carcinoma (54%), adenocarcinoma (25%), and mucoepidermoid carcinoma (13%). All 24 patients had an ETV6-NTRK3 gene fusion. The ORR was 92% (95% confidence interval, 73-99). Best overall response was complete response in three (13%) patients, partial response in 19 (79%), and progressive disease in two (8%). The rate of progression-free survival at 24 months was 78% (median follow-up 30.9 months). Most treatment-related adverse events (AEs) were grade 1-2, and no patients discontinued treatment due to AEs. CONCLUSION Larotrectinib demonstrated robust and durable efficacy in patients with TRK fusion-positive salivary gland tumors of various histologies, and a favorable safety profile. These findings support NTRK gene fusion testing in patients with advanced salivary gland cancers. CLINICALTRIALS.GOV NUMBERS NCT02122913 and NCT02576431.
Collapse
Affiliation(s)
- Xiuning Le
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina Baik
- University of Washington/Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Jessica Bauman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Marcia S Brose
- Sidney Kimmel Cancer Center of Jefferson University Health, Philadelphia, PA, USA
| | | | - Tejas Patil
- Department of Medicine, Division of Medical Oncology, University of Colorado, Denver, Aurora, CO, USA
| | - Ray McDermott
- St. Vincent’s University Hospital, Dublin, Ireland
- Cancer Trials Ireland, Dublin, Ireland
| | - Luis E Raez
- Memorial Cancer Institute, Florida International University, Miami, FL, USA
| | - Jennifer M Johnson
- Sidney Kimmel Cancer Center of Jefferson University Health, Philadelphia, PA, USA
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Makoto Tahara
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Alan L Ho
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | - Laura Dima
- Bayer HealthCare Pharmaceuticals, Inc., Basel, Switzerland
| | | | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - David S Hong
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
147
|
Kelly GA, Venkatramani R, Quintanilla NM, Chelius DC, Roy A, Mahajan P. Secretory Carcinoma of the Salivary Gland: A Rarity in Children. J Pediatr Hematol Oncol 2022; 44:167-172. [PMID: 34486543 DOI: 10.1097/mph.0000000000002304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/29/2021] [Indexed: 11/27/2022]
Abstract
Originally described as mammary analog secretory carcinoma (SC), SC of the salivary gland is a rare malignancy with morphologic and molecular similarities to SC of the breast. We present 2 children with salivary gland SC with the classic ETV6-NTRK3 gene fusion, including 1 with lymph node metastases. Both patients underwent surgical resection and were in remission 24 months postsurgery. One patient was additionally found to have synchronous papillary thyroid carcinoma with a TFG-MET fusion. A review of published cases highlights the expanding molecular profile and confirms the favorable course of salivary gland SC after surgical resection.
Collapse
Affiliation(s)
| | - Rajkumar Venkatramani
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children's Cancer Center
| | | | - Daniel C Chelius
- Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX
| | - Angshumoy Roy
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children's Cancer Center
- Departments of Pathology and Immunology
| | - Priya Mahajan
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children's Cancer Center
| |
Collapse
|
148
|
Malla M, Parikh AR. Evolving Role of Circulating Tumor DNA and Emerging Targeted Therapy in Colorectal Cancer. Hematol Oncol Clin North Am 2022; 36:583-601. [DOI: 10.1016/j.hoc.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
149
|
Moes-Sosnowska J, Chorostowska-Wynimko J. Fibroblast Growth Factor Receptor 1-4 Genetic Aberrations as Clinically Relevant Biomarkers in Squamous Cell Lung Cancer. Front Oncol 2022; 12:780650. [PMID: 35402233 PMCID: PMC8991910 DOI: 10.3389/fonc.2022.780650] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor receptor (FGFR) inhibitors (FGFRis) are a potential therapeutic option for squamous non-small cell lung cancer (Sq-NSCLC). Because appropriate patient selection is needed for targeted therapy, molecular profiling is key to discovering candidate biomarker(s). Multiple FGFR aberrations are present in Sq-NSCLC tumors-alterations (mutations and fusions), amplification and mRNA/protein overexpression-but their predictive potential is unclear. Although FGFR1 amplification reliability was unsatisfactory, FGFR mRNA overexpression, mutations, and fusions are promising. However, currently their discriminatory power is insufficient, and the available clinical data are from small groups of Sq-NSCLC patients. Here, we focus on FGFR aberrations as predictive biomarkers for FGFR-targeting agents in Sq-NSCLC. Known and suggested molecular determinants of FGFRi resistance are also discussed.
Collapse
Affiliation(s)
- Joanna Moes-Sosnowska
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| |
Collapse
|
150
|
Chakravarty D, Johnson A, Sklar J, Lindeman NI, Moore K, Ganesan S, Lovly CM, Perlmutter J, Gray SW, Hwang J, Lieu C, André F, Azad N, Borad M, Tafe L, Messersmith H, Robson M, Meric-Bernstam F. Somatic Genomic Testing in Patients With Metastatic or Advanced Cancer: ASCO Provisional Clinical Opinion. J Clin Oncol 2022; 40:1231-1258. [PMID: 35175857 DOI: 10.1200/jco.21.02767] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE An ASCO provisional clinical opinion offers timely clinical direction to ASCO's membership following publication or presentation of potentially practice-changing data from major studies. This provisional clinical opinion addresses the appropriate use of tumor genomic testing in patients with metastatic or advanced solid tumors. CLINICAL CONTEXT An increasing number of therapies are approved to treat cancers harboring specific genomic biomarkers. However, there is a lack of clarity as to when tumor genomic sequencing should be ordered, what type of assays should be performed, and how to interpret the results for treatment selection. PROVISIONAL CLINICAL OPINION Patients with metastatic or advanced cancer should undergo genomic sequencing in a certified laboratory if the presence of one or more specific genomic alterations has regulatory approval as biomarkers to guide the use of or exclusion from certain treatments for their disease. Multigene panel-based assays should be used if more than one biomarker-linked therapy is approved for the patient's disease. Site-agnostic approvals for any cancer with a high tumor mutation burden, mismatch repair deficiency, or neurotrophic tyrosine receptor kinase (NTRK) fusions provide a rationale for genomic testing for all solid tumors. Multigene testing may also assist in treatment selection by identifying additional targets when there are few or no genotype-based therapy approvals for the patient's disease. For treatment planning, the clinician should consider the functional impact of the targeted alteration and expected efficacy of genomic biomarker-linked options relative to other approved or investigational treatments.Additional information is available at www.asco.org/assays-and-predictive-markers-guidelines.
Collapse
Affiliation(s)
| | | | | | - Neal I Lindeman
- Brigham and Womens' Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | - Fabrice André
- PRISM, Precision Medicine Center, Institut Gustave Roussy, Villejuif, France
| | | | | | - Laura Tafe
- Dartmouth-Hitchcock Medical Center and The Geisel School of Medicine at Dartmouth, Darmouth, NH
| | | | - Mark Robson
- Memorial Sloan Kettering Cancer Center, New York City, NY
| | | |
Collapse
|