1
|
Dietz MV, van Kooten JP, Paats MS, Aerts JGVJ, Verhoef C, Madsen EVE, Dubbink HJ, von der Thüsen JH. Molecular alterations and potential actionable mutations in peritoneal mesothelioma: a scoping review of high-throughput sequencing studies. ESMO Open 2023; 8:101600. [PMID: 37453150 PMCID: PMC10368826 DOI: 10.1016/j.esmoop.2023.101600] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Peritoneal mesothelioma (PeM) is a rare malignancy with a poor prognosis. Currently there is a lack of effective systemic therapies. Due to the rarity of PeM, it is challenging to study new treatment options. Off-label use of targeted drugs could be an effective approach. This scoping review aims to explore the genomic landscape of PeM to identify potential therapeutic targets. MATERIALS AND METHODS A systematic literature search of Embase, Medline, Web of Science, the Cochrane Library, and Google Scholar was carried out up to 1 November 2022. Studies that reported on molecular alterations in PeM detected by high-throughput sequencing techniques were included. Genes that were altered in ≥1% of PeMs were selected for the identification of potential targeted therapies. RESULTS Thirteen articles were included, comprising 824 PeM patients. In total, 142 genes were altered in ≥1% of patients, of which 7 genes were altered in ≥10%. BAP1 was the most commonly altered gene (50%). Other commonly altered genes were NF2 (25%), CDKN2A (23%), CDKN2B (17%), PBRM1 (15%), TP53 (14%), and SETD2 (13%). In total, 17% of PeM patients were carriers of a germline mutation, mainly in BAP1 (7%). CONCLUSIONS This scoping review provides an overview of the mutational landscape of PeM. Germline mutations might be a larger contributor to the incidence of PeM than previously thought. Currently available targeted therapy options are limited, but several targeted agents [such as poly (ADP-ribose) polymerase (PARP), enhancer of zeste homolog 2 (EZH2), and cyclin-dependent kinase 4/6 (CDK4/6) inhibitors] were identified that might provide new targeted therapy options in the future.
Collapse
Affiliation(s)
| | | | - M S Paats
- Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam
| | - J G V J Aerts
- Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam
| | | | | | - H J Dubbink
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
2
|
Lavoie JM, Csizmok V, Williamson LM, Culibrk L, Wang G, Marra MA, Laskin J, Jones SJM, Renouf DJ, Kollmannsberger CK. Whole-genome and transcriptome analysis of advanced adrenocortical cancer highlights multiple alterations affecting epigenome and DNA repair pathways. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006148. [PMID: 35483882 PMCID: PMC9059790 DOI: 10.1101/mcs.a006148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/18/2022] [Indexed: 12/20/2022] Open
Abstract
Adrenocortical cancer (ACC) is a rare cancer of the adrenal gland. Several driver mutations have been identified in both primary and metastatic ACCs, but the therapeutic options are still limited. We performed whole-genome and transcriptome sequencing on seven patients with metastatic ACC. Integrative analysis of mutations, RNA expression changes, mutation signature, and homologous recombination deficiency (HRD) analysis was performed. Mutations affecting CTNNB1 and TP53 and frequent loss of heterozygosity (LOH) events were observed in our cohort. Alterations affecting genes involved in cell cycle (RB1, CDKN2A, CDKN2B), DNA repair pathways (MUTYH, BRCA2, ATM, RAD52, MLH1, MSH6), and telomere maintenance (TERF2 and TERT) consisting of somatic and germline mutations, structural variants, and expression outliers were also observed. HRDetect, which aggregates six HRD-associated mutation signatures, identified a subset of cases as HRD. Genomic alterations affecting genes involved in epigenetic regulation were also identified, including structural variants (SWI/SNF genes and histone methyltransferases), and copy gains and concurrent high expression of KDM5A, which may contribute to epigenomic deregulation. Findings from this study highlight HRD and epigenomic pathways as potential therapeutic targets and suggest a subgroup of patients may benefit from a diverse array of molecularly targeted therapies in ACC, a rare disease in urgent need of therapeutic strategies.
Collapse
Affiliation(s)
- Jean-Michel Lavoie
- Department of Medical Oncology, BC Cancer, Surrey, British Columbia V3V 1Z2, Canada
| | - Veronika Csizmok
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | - Luka Culibrk
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | - Gang Wang
- Department of Pathology and Laboratory Medicine, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | - Daniel J Renouf
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia V5Z 4E6, Canada
| | | |
Collapse
|
3
|
Fortarezza F, Pezzuto F, Marzullo A, Cavone D, Romano DE, d'Amati A, Serio G, Vimercati L. Molecular Pathways in Peritoneal Mesothelioma: A Minireview of New Insights. Front Oncol 2022; 12:823839. [PMID: 35223506 PMCID: PMC8866824 DOI: 10.3389/fonc.2022.823839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
Mesothelioma is a rare malignant neoplasm with poor survival. It mainly affects the pleura (90%) but can arise in all serous cavities: peritoneum (5-10%), pericardium and tunica vaginalis testis (<1%). The onset of pleural mesothelioma is strictly related to asbestos exposure with a long latency time. The causal link with asbestos has also been suggested for peritoneal mesothelioma, while the importance of exposure in the onset of pericardial and tunica vaginalis testis mesotheliomas is not well known. Mesothelioma remains an aggressive and fatal disease with a five-year mortality rate higher than 95%. However, new therapeutic approaches based on molecular-targeted and immunomodulatory therapies are being explored but have conflicting results. In this context, the identification of critical targets appears mandatory. Awareness of the molecular and physiological changes leading to the neoplastic degeneration of mesothelial cells and the identification of gene mutations, epigenetic alterations, gene expression profiles and altered pathways could be helpful for selecting targetable mechanisms and molecules. In this review, we aimed to report recent research in the last 20 years focusing on the molecular pathways and prognostic factors in peritoneal mesothelioma and their possible diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Pathology Unit, University of Padova, Padova, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Pathology Unit, University of Padova, Padova, Italy
| | - Andrea Marzullo
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Domenica Cavone
- Department of Interdisciplinary Medicine, Occupational Health Unit, University of Bari, Bari, Italy
| | - Daniele Egidio Romano
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Antonio d'Amati
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Gabriella Serio
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Luigi Vimercati
- Department of Interdisciplinary Medicine, Occupational Health Unit, University of Bari, Bari, Italy
| |
Collapse
|
4
|
A rare case of malignant peritoneal mesothelioma with EWSR-ATF1 fusion transcription and unusual immunophenotype. HUMAN PATHOLOGY: CASE REPORTS 2021. [DOI: 10.1016/j.ehpc.2021.200542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
5
|
Hung YP, Dong F, Torre M, Crum CP, Bueno R, Chirieac LR. Molecular characterization of diffuse malignant peritoneal mesothelioma. Mod Pathol 2020; 33:2269-2279. [PMID: 32504035 DOI: 10.1038/s41379-020-0588-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/23/2020] [Accepted: 05/24/2020] [Indexed: 01/31/2023]
Abstract
Malignant peritoneal mesothelioma is a rare aggressive tumor that arises from the peritoneal lining. While recurrent BAP1 mutations have been identified in a subset of mesotheliomas, molecular characteristics of peritoneal mesotheliomas, including those lacking BAP1 alterations, remain poorly understood. Using targeted next-generation sequencing, we examined the molecular features of 26 diffuse malignant peritoneal mesotheliomas. As part of an exploratory analysis, we analyzed an additional localized peritoneal mesothelioma and one well-differentiated papillary mesothelioma with invasive foci. Genomic characterization identified categories of diffuse malignant peritoneal mesotheliomas: The first group included 18 (69%) tumors with recurrent BAP1 alterations, with eight (31%) having more than one BAP1 alterations, and concomitant alterations in PBRM1 (46%) and SETD2 (35%). All tumors with complete loss of BAP1 expression by immunohistochemistry harbored BAP1 molecular alterations. PBRM1 alterations were significantly enriched in the BAP1-altered cohort. Frequent copy number loss of BAP1, ARID1B, PRDM1, PBRM1, SETD2, NF2, and CDKN2A was noted. The second group included eight (31%) BAP1-wild-type tumors: two with TP53 mutations, one with a TRAF7 activating mutation, one with a SUZ12 inactivating mutation, and three with ALK rearrangements that we previously published. One TP53-mutant biphasic mesothelioma showed evidence of genomic near-haploidization showing loss of heterozygosity of all chromosomes except 5, 7, 16, and 20. The localized peritoneal mesothelioma harbored a nonsense CHEK2 mutation, and the well-differentiated papillary mesothelioma with invasive foci harbored no reportable variants. In conclusion, we described the genetic categories of diffuse malignant peritoneal mesotheliomas, with BAP1-mutant and BAP1-wild-type groups. Our findings implicated DNA repair, epigenetics, and cell cycle regulation in the pathogenesis of peritoneal mesotheliomas, with identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Yin P Hung
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA. .,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew Torre
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher P Crum
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Raphael Bueno
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lucian R Chirieac
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Mutational Profile of Malignant Pleural Mesothelioma (MPM) in the Phase II RAMES Study. Cancers (Basel) 2020; 12:cancers12102948. [PMID: 33065998 PMCID: PMC7601196 DOI: 10.3390/cancers12102948] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Malignant pleura Mesothelioma (MPM) is an aggressive cancer arising from the mesothelial cells of the pleura. About 80% of mesothelioma cases are linked to asbestos exposure; the remainder may be related to prior chest radiation, genetic predisposition or spontaneous occurrence. Understanding the genetic alterations that drive MPM is critical for successful development of diagnostics, prognostics and personalized therapeutic modalities. Because MPM is rare, genomic studies are limited and have typically involved a small number of samples. The aim of our study was to understand the mutational landscape of MPM. Our analysis identified significantly mutated genes. This study on a rare tumor type will be important for patients “in real life”. Abstract Purpose: Malignant pleural mesothelioma (MPM) is an aggressive cancer. Data are not available in prospective trials on correlations between genetic alterations and outcomes of therapies. In this study, we assessed the genetic profile of MPM patients (pts) in tissue samples. Patients and Methods: From December 2016 to July 2018 (end of enrolment), 164 pts were enrolled. We evaluated by targeted sequencing the mutational profile of a panel of 34 genes: ACTB, ACTG1, ACTG2, ACTR1A, BAP1, CDH8, CDK4, CDKN2A, CDKN2B, COL3A1, COL5A2, CUL1, DHFR, GOT1, KDR, KIT, MXRA5, NF2, NFRKB, NKX6-2, NOD2, PCBD2, PDZK1IP1, PIK3CA, PIK3CB, PSMD13, RAPGEF6, RDX, SETDB1, TAOK1, TP53, TXNRD1, UQCRC1, XRCC6. Genetic profiling was correlated with clinical and pathological variables. Results: Overall, 110 pts (67%) from both treatment arms had samples available for molecular analysis. Median age was 63 years (45–81), 25.5% (n = 28) were females, and 74.5% (n = 82) were males. Tumor histotype was 81.8% (n = 90) epithelioid and 18.2% (n = 20) non-epithelioid; 28.5% of the tumors (n = 42) were stage IV, 71.5% (n = 68) were stage III. Targeted sequencing of tissue specimens identified 275 functional somatic mutations in the 34 genes analyzed. The number of mutated genes was positively associated with higher stage and metastatic disease (p = 0.025). RDX (42%), MXRA5 (23%), BAP1 (14%), and NF2 (11%) were the most frequently mutated genes. Mutations in RAPGEF6 (p = 0.03) and ACTG1 (p = 0.02) were associated with the non-epithelioid subtype, and mutations in BAP1 (p = 0.04) were related to progression-free survival (PFS) > 6 months. Conclusions: In the Ramucirumab Mesothelioma clinical trial (RAMES), mutation of the gene BAP1 is related to a prolonged PFS for patients treated with platinum/pemetrexed regimens (p = 0.04).
Collapse
|
7
|
Rodchenkov I, Babur O, Luna A, Aksoy BA, Wong JV, Fong D, Franz M, Siper MC, Cheung M, Wrana M, Mistry H, Mosier L, Dlin J, Wen Q, O’Callaghan C, Li W, Elder G, Smith PT, Dallago C, Cerami E, Gross B, Dogrusoz U, Demir E, Bader GD, Sander C. Pathway Commons 2019 Update: integration, analysis and exploration of pathway data. Nucleic Acids Res 2020; 48:D489-D497. [PMID: 31647099 PMCID: PMC7145667 DOI: 10.1093/nar/gkz946] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Pathway Commons (https://www.pathwaycommons.org) is an integrated resource of publicly available information about biological pathways including biochemical reactions, assembly of biomolecular complexes, transport and catalysis events and physical interactions involving proteins, DNA, RNA, and small molecules (e.g. metabolites and drug compounds). Data is collected from multiple providers in standard formats, including the Biological Pathway Exchange (BioPAX) language and the Proteomics Standards Initiative Molecular Interactions format, and then integrated. Pathway Commons provides biologists with (i) tools to search this comprehensive resource, (ii) a download site offering integrated bulk sets of pathway data (e.g. tables of interactions and gene sets), (iii) reusable software libraries for working with pathway information in several programming languages (Java, R, Python and Javascript) and (iv) a web service for programmatically querying the entire dataset. Visualization of pathways is supported using the Systems Biological Graphical Notation (SBGN). Pathway Commons currently contains data from 22 databases with 4794 detailed human biochemical processes (i.e. pathways) and ∼2.3 million interactions. To enhance the usability of this large resource for end-users, we develop and maintain interactive web applications and training materials that enable pathway exploration and advanced analysis.
Collapse
Affiliation(s)
- Igor Rodchenkov
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Ozgun Babur
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Augustin Luna
- cBio Center, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Bulent Arman Aksoy
- Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, New York, NY 10065, USA
| | - Jeffrey V Wong
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Dylan Fong
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Max Franz
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Metin Can Siper
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Manfred Cheung
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Michael Wrana
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Harsh Mistry
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Logan Mosier
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Jonah Dlin
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Qizhi Wen
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Caitlin O’Callaghan
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Wanxin Li
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Geoffrey Elder
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Peter T Smith
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Christian Dallago
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02215, USA
- Department of Informatics, Technische Universität München, 85748 Garching, Germany
| | - Ethan Cerami
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Benjamin Gross
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ugur Dogrusoz
- Department of Computer Engineering, Bilkent University, Ankara 06800, Turkey
| | - Emek Demir
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Chris Sander
- cBio Center, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
8
|
Feng X, Pleasance E, Zhao EY, Ng T, Grewal JK, Mohammad N, Taylor SK, Simmons C, Srikanthan A, Rassekh SR, Deyell R, Rauw J, Knowling M, Khoo K, Lee U, Noonan K, Hart J, Tonseth RP, Shen Y, Titmuss E, Jones M, Bonakdar M, Reisle C, Taylor GA, Chan S, Mungall K, Chuah E, Zhao Y, Mungall A, Moore R, Lim H, Renouf DJ, Gelmon K, Yip S, Jones SJM, Marra M, Laskin J. Therapeutic Implication of Genomic Landscape of Adult Metastatic Sarcoma. JCO Precis Oncol 2019; 3:1-25. [PMID: 35100702 DOI: 10.1200/po.18.00325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE This study investigated therapeutic potential of integrated genome and transcriptome profiling of metastatic sarcoma, a rare but extremely heterogeneous group of aggressive mesenchymal malignancies with few systemic therapeutic options. METHODS Forty-three adult patients with advanced or metastatic non-GI stromal tumor sarcomas of various histology subtypes who were enrolled in the Personalized OncoGenomics program at BC Cancer were included in this study. Fresh tumor tissues along with blood samples underwent whole-genome and transcriptome sequencing. RESULTS The most frequent genomic alterations in this cohort are large-scale structural variation and somatic copy number variation. Outlier RNA expression as well as somatic copy number variations, structural variations, and small mutations together suggest the presence of one or more potential therapeutic targets in the majority of patients in our cohort. Point mutations or deletions in known targetable cancer genes are rare; for example, tuberous sclerosis complex 2 provides a rationale for targeting the mammalian target of rapamycin pathway, resulting in a few patients with exceptional clinical benefit from everolimus. In addition, we observed recurrent 17p11-12 amplifications, which seem to be a sarcoma-specific event. This may suggest that this region harbors an oncogene(s) that is significant for sarcoma tumorigenesis. Furthermore, some sarcoma tumors carrying a distinct mutational signature suggestive of homologous recombination deficiency seem to demonstrate sensitivity to double-strand DNA-damaging agents. CONCLUSION Integrated large-scale genomic analysis may provide insights into potential therapeutic targets as well as novel biologic features of metastatic sarcomas that could fuel future experimental and clinical research and help design biomarker-driven basket clinical trials for novel therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Eric Y Zhao
- BC Cancer, Vancouver, British Columbia, Canada
| | - Tony Ng
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Sara K Taylor
- BC Cancer-Kelowna, Kelowna, British Columbia, Canada
| | | | | | - S Rod Rassekh
- BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Rebecca Deyell
- BC Children's Hospital, Vancouver, British Columbia, Canada
| | | | | | - Kong Khoo
- BC Cancer, Surrey, British Columbia, Canada
| | - Ursula Lee
- BC Cancer, Surrey, British Columbia, Canada
| | | | - Jason Hart
- BC Cancer, Victoria, British Columbia, Canada
| | | | | | | | | | | | | | | | - Simon Chan
- BC Cancer, Vancouver, British Columbia, Canada
| | | | - Eric Chuah
- BC Cancer, Vancouver, British Columbia, Canada
| | | | | | | | - Howard Lim
- BC Cancer, Vancouver, British Columbia, Canada
| | | | | | - Stephen Yip
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Marco Marra
- BC Cancer, Vancouver, British Columbia, Canada
| | | |
Collapse
|
9
|
Genomic characterization of a well-differentiated grade 3 pancreatic neuroendocrine tumor. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a003814. [PMID: 31160355 PMCID: PMC6549554 DOI: 10.1101/mcs.a003814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/06/2019] [Indexed: 01/05/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) represent a minority of pancreatic neoplasms that exhibit variability in prognosis. Ongoing mutational analyses of PanNENs have found recurrent abnormalities in chromatin remodeling genes (e.g., DAXX and ATRX), and mTOR pathway genes (e.g., TSC2, PTEN PIK3CA, and MEN1), some of which have relevance to patients with related familial syndromes. Most recently, grade 3 PanNENs have been divided into two groups based on differentiation, creating a new group of well-differentiated grade 3 neuroendocrine tumors (PanNETs) that have had a limited whole-genome level characterization to date. In a patient with a metastatic well-differentiated grade 3 PanNET, our study utilized whole-genome sequencing of liver metastases for the comparative analysis and detection of single-nucleotide variants, insertions and deletions, structural variants, and copy-number variants, with their biologic relevance confirmed by RNA sequencing. We found that this tumor most notably exhibited a TSC1-disrupting fusion, showed a novel CHD7–BEND2 fusion, and lacked any somatic variants in ATRX, DAXX, and MEN1.
Collapse
|
10
|
Integrative genomic analysis of peritoneal malignant mesothelioma: understanding a case with extraordinary chemotherapy response. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a003566. [PMID: 30862609 PMCID: PMC6549577 DOI: 10.1101/mcs.a003566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/17/2019] [Indexed: 12/31/2022] Open
Abstract
Peritoneal malignant mesothelioma is a rare disease with a generally poor prognosis and poor response to chemotherapy. To improve survival there is a need for increased molecular understanding of the disease, including chemotherapy sensitivity and resistance. We here present an unusual case concerning a young woman with extensive peritoneal mesothelioma who had a remarkable response to palliative chemotherapy (platinum/pemetrexed). Tumor samples collected at surgery before and after treatment were analyzed on the genomic and transcriptional levels (exome sequencing, RNA-seq, and smallRNA-seq). Integrative analysis of single nucleotide and copy-number variants, mutational signatures, and gene expression was performed to provide a comprehensive picture of the disease. LATS1/2 were identified as the main mutational drivers together with homozygous loss of BAP1 and PBRM1, which also may have contributed to the extraordinary chemotherapy response. The presence of the S3 mutational signature is consistent with homologous recombination DNA repair defects due to BAP1 loss. Up-regulation of the PI3K/AKT/mTOR pathway after treatment, supported by deactivated PTEN through miRNA regulation, is associated with cancer progression and could explain chemotherapy resistance. The molecular profile suggests potential benefit from experimental targeting of PARP, EZH2, the PI3K/AKT/mTOR pathway and possibly also from immune checkpoint inhibition. In addition to providing the molecular background for this unusual case of peritoneal mesothelioma, the results show the potential value of integrative genomic analysis in precision medicine.
Collapse
|
11
|
Shrestha R, Nabavi N, Lin YY, Mo F, Anderson S, Volik S, Adomat HH, Lin D, Xue H, Dong X, Shukin R, Bell RH, McConeghy B, Haegert A, Brahmbhatt S, Li E, Oo HZ, Hurtado-Coll A, Fazli L, Zhou J, McConnell Y, McCart A, Lowy A, Morin GB, Chen T, Daugaard M, Sahinalp SC, Hach F, Le Bihan S, Gleave ME, Wang Y, Churg A, Collins CC. BAP1 haploinsufficiency predicts a distinct immunogenic class of malignant peritoneal mesothelioma. Genome Med 2019; 11:8. [PMID: 30777124 PMCID: PMC6378747 DOI: 10.1186/s13073-019-0620-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background Malignant peritoneal mesothelioma (PeM) is a rare and fatal cancer that originates from the peritoneal lining of the abdomen. Standard treatment of PeM is limited to cytoreductive surgery and/or chemotherapy, and no effective targeted therapies for PeM exist. Some immune checkpoint inhibitor studies of mesothelioma have found positivity to be associated with a worse prognosis. Methods To search for novel therapeutic targets for PeM, we performed a comprehensive integrative multi-omics analysis of the genome, transcriptome, and proteome of 19 treatment-naïve PeM, and in particular, we examined BAP1 mutation and copy number status and its relationship to immune checkpoint inhibitor activation. Results We found that PeM could be divided into tumors with an inflammatory tumor microenvironment and those without and that this distinction correlated with haploinsufficiency of BAP1. To further investigate the role of BAP1, we used our recently developed cancer driver gene prioritization algorithm, HIT’nDRIVE, and observed that PeM with BAP1 haploinsufficiency form a distinct molecular subtype characterized by distinct gene expression patterns of chromatin remodeling, DNA repair pathways, and immune checkpoint receptor activation. We demonstrate that this subtype is correlated with an inflammatory tumor microenvironment and thus is a candidate for immune checkpoint blockade therapies. Conclusions Our findings reveal BAP1 to be a potential, easily trackable prognostic and predictive biomarker for PeM immunotherapy that refines PeM disease classification. BAP1 stratification may improve drug response rates in ongoing phases I and II clinical trials exploring the use of immune checkpoint blockade therapies in PeM in which BAP1 status is not considered. This integrated molecular characterization provides a comprehensive foundation for improved management of a subset of PeM patients. Electronic supplementary material The online version of this article (10.1186/s13073-019-0620-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raunak Shrestha
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Bioinformatics Training Program, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Noushin Nabavi
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Fan Mo
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,International Precision Medicine Research Centre, Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China.,Neoantigen Therapeutics, Inc., Hangzhou, 310051, Zhejiang, China
| | - Shawn Anderson
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Stanislav Volik
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Hans H Adomat
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Dong Lin
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Hui Xue
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Xin Dong
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Robert Shukin
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Robert H Bell
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Brian McConeghy
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Sonal Brahmbhatt
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Estelle Li
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | | | - Ladan Fazli
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Joshua Zhou
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Yarrow McConnell
- Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Andrea McCart
- Mount Sinai Hospital, 600 University Ave, Toronto, ON, M5G 1X5, Canada
| | - Andrew Lowy
- Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA
| | - Gregg B Morin
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Tianhui Chen
- Zhejiang Academy of Medical Sciences, Tianmushan Road 182, Hangzhou, 310013, China
| | - Mads Daugaard
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - S Cenk Sahinalp
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,School of Informatics and Computing, Indiana University, Bloomington, IN, 47408, USA
| | - Faraz Hach
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Stephane Le Bihan
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Andrew Churg
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada.
| | - Colin C Collins
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
12
|
A high-throughput protocol for isolating cell-free circulating tumor DNA from peripheral blood. Biotechniques 2019; 66:85-92. [PMID: 30744412 DOI: 10.2144/btn-2018-0148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The analysis of cell-free circulating tumor DNA (ctDNA) is potentially a less invasive, more dynamic assessment of cancer progression and treatment response than characterizing solid tumor biopsies. Standard isolation methods require separation of plasma by centrifugation, a time-consuming step that complicates automation. To address these limitations, we present an automatable magnetic bead-based ctDNA isolation method that eliminates centrifugation to purify ctDNA directly from peripheral blood (PB). To develop and test our method, ctDNA from cancer patients was purified from PB and plasma. We found that allelic fractions of somatic single-nucleotide variants from target gene capture libraries were comparable, indicating that the PB ctDNA purification method may be a suitable replacement for the plasma-based protocols currently in use.
Collapse
|
13
|
Wong HL, Zhao EY, Jones MR, Reisle CR, Eirew P, Pleasance E, Grande BM, Karasinska JM, Kalloger SE, Lim HJ, Shen Y, Yip S, Morin RD, Laskin J, Marra MA, Jones SJ, Schrader KA, Schaeffer DF, Renouf DJ. Temporal Dynamics of Genomic Alterations in a BRCA1 Germline-Mutated Pancreatic Cancer With Low Genomic Instability Burden but Exceptional Response to Fluorouracil, Oxaliplatin, Leucovorin, and Irinotecan. JCO Precis Oncol 2018; 2:PO.18.00057. [PMID: 32913994 PMCID: PMC7446469 DOI: 10.1200/po.18.00057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Hui-li Wong
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Eric Y. Zhao
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Martin R. Jones
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Caralyn R. Reisle
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Peter Eirew
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Erin Pleasance
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Bruno M. Grande
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Joanna M. Karasinska
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Steve E. Kalloger
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Howard J. Lim
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Yaoqing Shen
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Stephen Yip
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Ryan D. Morin
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Janessa Laskin
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Marco A. Marra
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Steven J.M. Jones
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Kasmintan A. Schrader
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - David F. Schaeffer
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Daniel J. Renouf
- Hui-li Wong, Eric Y. Zhao, Martin R. Jones, Caralyn R. Reisle, Peter Eirew, Erin Pleasance, Howard J. Lim, Yaoqing Shen, Ryan D. Morin, Janessa Laskin, Marco A. Marra, Steven J.M. Jones, and Daniel J. Renouf, British Columbia Cancer Agency; Hui-li Wong, Joanna M. Karasinska, Steve E. Kalloger, David F. Schaeffer, and Daniel J. Renouf, Pancreas Centre BC; Bruno M. Grande and Ryan D. Morin, Simon Fraser University; Steve E. Kalloger, Stephen Yip, Marco A. Marra, Steven J.M. Jones, Kasmintan A. Schrader, and David F. Schaeffer, University of British Columbia; and David F. Schaeffer, Vancouver General Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
14
|
Clinical cancer genomic profiling by three-platform sequencing of whole genome, whole exome and transcriptome. Nat Commun 2018; 9:3962. [PMID: 30262806 PMCID: PMC6160438 DOI: 10.1038/s41467-018-06485-7] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/24/2018] [Indexed: 12/17/2022] Open
Abstract
To evaluate the potential of an integrated clinical test to detect diverse classes of somatic and germline mutations relevant to pediatric oncology, we performed three-platform whole-genome (WGS), whole exome (WES) and transcriptome (RNA-Seq) sequencing of tumors and normal tissue from 78 pediatric cancer patients in a CLIA-certified, CAP-accredited laboratory. Our analysis pipeline achieves high accuracy by cross-validating variants between sequencing types, thereby removing the need for confirmatory testing, and facilitates comprehensive reporting in a clinically-relevant timeframe. Three-platform sequencing has a positive predictive value of 97–99, 99, and 91% for somatic SNVs, indels and structural variations, respectively, based on independent experimental verification of 15,225 variants. We report 240 pathogenic variants across all cases, including 84 of 86 known from previous diagnostic testing (98% sensitivity). Combined WES and RNA-Seq, the current standard for precision oncology, achieved only 78% sensitivity. These results emphasize the critical need for incorporating WGS in pediatric oncology testing. Clinical oncology is rapidly adopting next-generation sequencing technology for nucleotide variant and indel detection. Here the authors present a three-platform approach (whole-genome, whole-exome, and whole-transcriptome) in pediatric patients for the detection of diverse types of germline and somatic variants.
Collapse
|
15
|
Zhao EY, Shen Y, Pleasance E, Kasaian K, Leelakumari S, Jones M, Bose P, Ch'ng C, Reisle C, Eirew P, Corbett R, Mungall KL, Thiessen N, Ma Y, Schein JE, Mungall AJ, Zhao Y, Moore RA, Den Brok W, Wilson S, Villa D, Shenkier T, Lohrisch C, Chia S, Yip S, Gelmon K, Lim H, Renouf D, Sun S, Schrader KA, Young S, Bosdet I, Karsan A, Laskin J, Marra MA, Jones SJM. Homologous Recombination Deficiency and Platinum-Based Therapy Outcomes in Advanced Breast Cancer. Clin Cancer Res 2018; 23:7521-7530. [PMID: 29246904 DOI: 10.1158/1078-0432.ccr-17-1941] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/14/2017] [Accepted: 09/26/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Recent studies have identified mutation signatures of homologous recombination deficiency (HRD) in over 20% of breast cancers, as well as pancreatic, ovarian, and gastric cancers. There is an urgent need to understand the clinical implications of HRD signatures. Whereas BRCA1/2 mutations confer sensitivity to platinum-based chemotherapies, it is not yet clear whether mutation signatures can independently predict platinum response.Experimental Design: In this observational study, we sequenced tumor whole genomes (100× depth) and matched normals (60×) of 93 advanced-stage breast cancers (33 platinum-treated). We computed a published metric called HRDetect, independently trained to predict BRCA1/2 status, and assessed its capacity to predict outcomes on platinum-based chemotherapies. Clinical endpoints were overall survival (OS), total duration on platinum-based therapy (TDT), and radiographic evidence of clinical improvement (CI).Results: HRDetect predicted BRCA1/2 status with an area under the curve (AUC) of 0.94 and optimal threshold of 0.7. Elevated HRDetect was also significantly associated with CI on platinum-based therapy (AUC = 0.89; P = 0.006) with the same optimal threshold, even after adjusting for BRCA1/2 mutation status and treatment timing. HRDetect scores over 0.7 were associated with a 3-month extended median TDT (P = 0.0003) and 1.3-year extended median OS (P = 0.04).Conclusions: Our findings not only independently validate HRDetect, but also provide the first evidence of its association with platinum response in advanced breast cancer. We demonstrate that HRD mutation signatures may offer clinically relevant information independently of BRCA1/2 mutation status and hope this work will guide the development of clinical trials. Clin Cancer Res; 23(24); 7521-30. ©2017 AACR.
Collapse
Affiliation(s)
- Eric Y Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Sreeja Leelakumari
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Martin Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Pinaki Bose
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Carolyn Ch'ng
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Caralyn Reisle
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Peter Eirew
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Richard Corbett
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Nina Thiessen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Yussanne Ma
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Jacqueline E Schein
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Wendie Den Brok
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Sheridan Wilson
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Diego Villa
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Tamara Shenkier
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Caroline Lohrisch
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Stephen Chia
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Gelmon
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Howard Lim
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Daniel Renouf
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Sophie Sun
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Kasmintan A Schrader
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Sean Young
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian Bosdet
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Aly Karsan
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Janessa Laskin
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada. .,Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Warren JSA, Xiao Y, Lamar JM. YAP/TAZ Activation as a Target for Treating Metastatic Cancer. Cancers (Basel) 2018; 10:cancers10040115. [PMID: 29642615 PMCID: PMC5923370 DOI: 10.3390/cancers10040115] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ) have both emerged as important drivers of cancer progression and metastasis. YAP and TAZ are often upregulated or nuclear localized in aggressive human cancers. There is abundant experimental evidence demonstrating that YAP or TAZ activation promotes cancer formation, tumor progression, and metastasis. In this review we summarize the evidence linking YAP/TAZ activation to metastasis, and discuss the roles of YAP and TAZ during each step of the metastatic cascade. Collectively, this evidence strongly suggests that inappropriate YAP or TAZ activity plays a causal role in cancer, and that targeting aberrant YAP/TAZ activation is a promising strategy for the treatment of metastatic disease. To this end, we also discuss several potential strategies for inhibiting YAP/TAZ activation in cancer and the challenges each strategy poses.
Collapse
Affiliation(s)
- Janine S A Warren
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
17
|
Kim JE, Kim D, Hong YS, Kim KP, Yoon YK, Lee DH, Kim SW, Chun SM, Jang SJ, Kim TW. Mutational Profiling of Malignant Mesothelioma Revealed Potential Therapeutic Targets in EGFR and NRAS. Transl Oncol 2018; 11:268-274. [PMID: 29413759 PMCID: PMC5884183 DOI: 10.1016/j.tranon.2018.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 12/29/2022] Open
Abstract
Pemetrexed and platinum (PP) combination chemotherapy is the current standard first-line therapy for treatment of malignant mesothelioma (MM). However, a useful predictive biomarker for PP therapy is yet to be found. Here, we performed targeted exome sequencing to profile somatic mutations and copy number variations in 12 MM patients treated with PP therapy. We identified 187 somatic mutations in 12 patients (65 synonymous, 102 missense, 2 nonsense, 5 splice site, and 13 small coding insertions/deletions). We identified somatic mutations in 23 genes including BAP1, TP53, NRAS, and EGFR. Interestingly, rare NRAS p.Q61K and EGFR exon 19 deletions were observed in 2 patients. We also found somatic chromosomal copy number deletions in CDKN2A and CDKN2B genes. Genetic alteration related to response after PP therapy was not found. Somatic mutation profiling in MM patients receiving PP therapy revealed genetic alterations in potential therapeutic targets such as NRAS and EGFR. No alterations in genes with potential predictive role for PP therapy were found.
Collapse
Affiliation(s)
- Jeong Eun Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul. Korea
| | - Deokhoon Kim
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea; Department of Pathology, University of Ulsan college of Medicine, Asan Medical Center, Seoul, Korea
| | - Yong Sang Hong
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul. Korea
| | - Kyu-Pyo Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul. Korea
| | - Young Kwang Yoon
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Dae Ho Lee
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul. Korea
| | - Sang-We Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul. Korea
| | - Sung-Min Chun
- Department of Pathology, University of Ulsan college of Medicine, Asan Medical Center, Seoul, Korea
| | - Se Jin Jang
- Department of Pathology, University of Ulsan college of Medicine, Asan Medical Center, Seoul, Korea
| | - Tae Won Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul. Korea.
| |
Collapse
|
18
|
Wong HL, Yang KC, Shen Y, Zhao EY, Loree JM, Kennecke HF, Kalloger SE, Karasinska JM, Lim HJ, Mungall AJ, Feng X, Davies JM, Schrader K, Zhou C, Karsan A, Jones SJM, Laskin J, Marra MA, Schaeffer DF, Gorski SM, Renouf DJ. Molecular characterization of metastatic pancreatic neuroendocrine tumors (PNETs) using whole-genome and transcriptome sequencing. Cold Spring Harb Mol Case Stud 2018; 4:mcs.a002329. [PMID: 29092957 PMCID: PMC5793777 DOI: 10.1101/mcs.a002329] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PNETs) are a genomically and clinically heterogeneous group of pancreatic neoplasms often diagnosed with distant metastases. Recurrent somatic mutations, chromosomal aberrations, and gene expression signatures in PNETs have been described, but the clinical significance of these molecular changes is still poorly understood, and the clinical outcomes of PNET patients remain highly variable. To help identify the molecular factors that contribute to PNET progression and metastasis, and as part of an ongoing clinical trial at the BC Cancer Agency (clinicaltrials.gov ID: NCT02155621), the genomic and transcriptomic profiles of liver metastases from five patients (four PNETs and one neuroendocrine carcinoma) were analyzed. In four of the five cases, we identified biallelic loss of MEN1 and DAXX as well as recurrent regions with loss of heterozygosity. Several novel findings were observed, including focal amplification of MYCN concomitant with loss of APC and TP53 in one sample with wild-type MEN1 and DAXX. Transcriptome analyses revealed up-regulation of MYCN target genes in this sample, confirming a MYCN-driven gene expression signature. We also identified a germline NTHL1 fusion event in one sample that resulted in a striking C>T mutation signature profile not previously reported in PNETs. These varying molecular alterations suggest different cellular pathways may contribute to PNET progression, consistent with the heterogeneous clinical nature of this disease. Furthermore, genomic profiles appeared to correlate well with treatment response, lending support to the role of prospective genotyping efforts to guide therapy in PNETs.
Collapse
Affiliation(s)
- Hui-Li Wong
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada.,Pancreas Centre BC, Vancouver, British Columbia V5Z 4E6, Canada
| | - Kevin C Yang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Vancouver, British Columbia V5A 1S6, Canada.,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Eric Y Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Jonathan M Loree
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Hagen F Kennecke
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Steve E Kalloger
- Pancreas Centre BC, Vancouver, British Columbia V5Z 4E6, Canada.,Division of Anatomical Pathology, Vancouver General Hospital, Vancouver, British Columbia V5Z 1M9, Canada
| | | | - Howard J Lim
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Xiaolan Feng
- Vancouver Island Centre, British Columbia Cancer Agency, Vancouver, British Columbia V8R 6V5, Canada
| | - Janine M Davies
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Kasmintan Schrader
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Chen Zhou
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Aly Karsan
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Steven J M Jones
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Vancouver, British Columbia V5A 1S6, Canada.,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Janessa Laskin
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - David F Schaeffer
- Pancreas Centre BC, Vancouver, British Columbia V5Z 4E6, Canada.,Division of Anatomical Pathology, Vancouver General Hospital, Vancouver, British Columbia V5Z 1M9, Canada
| | - Sharon M Gorski
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Vancouver, British Columbia V5A 1S6, Canada.,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Daniel J Renouf
- Division of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada.,Pancreas Centre BC, Vancouver, British Columbia V5Z 4E6, Canada
| |
Collapse
|
19
|
Sharif AA, Hergovich A. The NDR/LATS protein kinases in immunology and cancer biology. Semin Cancer Biol 2018; 48:104-114. [PMID: 28579171 DOI: 10.1016/j.semcancer.2017.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/15/2017] [Accepted: 04/25/2017] [Indexed: 02/07/2023]
|
20
|
Serio G, Pezzuto F, Marzullo A, Scattone A, Cavone D, Punzi A, Fortarezza F, Gentile M, Buonadonna AL, Barbareschi M, Vimercati L. Peritoneal Mesothelioma with Residential Asbestos Exposure. Report of a Case with Long Survival (Seventeen Years) Analyzed by Cgh-Array. Int J Mol Sci 2017; 18:E1818. [PMID: 28829357 PMCID: PMC5578204 DOI: 10.3390/ijms18081818] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/18/2022] Open
Abstract
Malignant mesothelioma is a rare and aggressive tumor with limited therapeutic options. We report a case of a malignant peritoneal mesothelioma (MPM) epithelioid type, with environmental asbestos exposure, in a 36-year-old man, with a long survival (17 years). The patient received standard treatment which included cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). METHODS AND RESULTS Molecular analysis with comparative genomic hybridization (CGH)-array was performed on paraffin-embedded tumoral samples. Multiple chromosomal imbalances were detected. The gains were prevalent. Losses at 1q21, 2q11.1→q13, 8p23.1, 9p12→p11, 9q21.33→q33.1, 9q12→q21.33, and 17p12→p11.2 are observed. Chromosome band 3p21 (BAP1), 9p21 (CDKN2A) and 22q12 (NF2) are not affected. Conclusions: the defects observed in this case are uncommon in malignant peritoneal mesothelioma. Some chromosomal aberrations that appear to be random here, might actually be relevant events explaining the response to therapy, the long survival and, finally, may be considered useful prognostic factors in peritoneal malignant mesothelioma (PMM).
Collapse
Affiliation(s)
- Gabriella Serio
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Federica Pezzuto
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Andrea Marzullo
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Anna Scattone
- Division of Pathology, IRCCS, National Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy.
| | - Domenica Cavone
- Department of Interdisciplinary Medicine, Occupational Health Division, Medical School, University of Bari, 70124 Bari, Italy.
| | - Alessandra Punzi
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Francesco Fortarezza
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Mattia Gentile
- Division of Medical Genetics "Di Venere Hospital", 70124 Bari, Italy.
| | | | | | - Luigi Vimercati
- Department of Interdisciplinary Medicine, Occupational Health Division, Medical School, University of Bari, 70124 Bari, Italy.
| |
Collapse
|
21
|
Prawira A, Pugh T, Stockley T, Siu L. Data resources for the identification and interpretation of actionable mutations by clinicians. Ann Oncol 2017; 28:946-957. [DOI: 10.1093/annonc/mdx023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
22
|
Sheffield BS, Tessier-Cloutier B, Li-Chang H, Shen Y, Pleasance E, Kasaian K, Li Y, Jones SJM, Lim HJ, Renouf DJ, Huntsman DG, Yip S, Laskin J, Marra M, Schaeffer DF. Personalized oncogenomics in the management of gastrointestinal carcinomas-early experiences from a pilot study. ACTA ACUST UNITED AC 2016; 23:e571-e575. [PMID: 28050146 DOI: 10.3747/co.23.3165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Gastrointestinal carcinomas are genomically complex cancers that are lethal in the metastatic setting. Whole-genome and transcriptome sequencing allow for the simultaneous characterization of multiple oncogenic pathways. METHODS We report 3 cases of metastatic gastrointestinal carcinoma in patients enrolled in the Personalized Onco-Genomics program at the BC Cancer Agency. Real-time genomic profiling was combined with clinical expertise to diagnose a carcinoma of unknown primary, to explore treatment response to bevacizumab in a colorectal cancer, and to characterize an appendiceal adenocarcinoma. RESULTS In the first case, genomic profiling revealed an IDH1 somatic mutation, supporting the diagnosis of cholangiocarcinoma in a malignancy of unknown origin, and further guided therapy by identifying epidermal growth factor receptor amplification. In the second case, a BRAF V600E mutation and wild-type KRAS profile justified the use of targeted therapies to treat a colonic adenocarcinoma. The third case was an appendiceal adenocarcinoma defined by a p53 inactivation; Ras/raf/mek, Akt/mtor, Wnt, and notch pathway activation; and overexpression of ret, erbb2 (her2), erbb3, met, and cell cycle regulators. SUMMARY We show that whole-genome and transcriptome sequencing can be achieved within clinically effective timelines, yielding clinically useful and actionable information.
Collapse
Affiliation(s)
- B S Sheffield
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - B Tessier-Cloutier
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - H Li-Chang
- Royal Victoria Regional Health Centre, Department of Pathology and Laboratory Medicine, Barrie, ON
| | - Y Shen
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - E Pleasance
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - K Kasaian
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - Y Li
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - S J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - H J Lim
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC
| | - D J Renouf
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC
| | - D G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - S Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - J Laskin
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC
| | - M Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC.; Department of Medical Genetics, University of British Columbia, Vancouver, BC
| | - D F Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| |
Collapse
|
23
|
Kang HC, Kim HK, Lee S, Mendez P, Kim JW, Woodard G, Yoon JH, Jen KY, Fang LT, Jones K, Jablons DM, Kim IJ. Whole exome and targeted deep sequencing identify genome-wide allelic loss and frequent SETDB1 mutations in malignant pleural mesotheliomas. Oncotarget 2016; 7:8321-31. [PMID: 26824986 PMCID: PMC4884995 DOI: 10.18632/oncotarget.7032] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/15/2016] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM), a rare malignancy with a poor prognosis, is mainly caused by exposure to asbestos or other organic fibers, but the underlying genetic mechanism is not fully understood. Genetic alterations and causes for multiple primary cancer development including MPM are unknown. We used whole exome sequencing to identify somatic mutations in a patient with MPM and two additional primary cancers who had no evidence of venous, arterial, lymphovascular, or perineural invasion indicating dissemination of a primary lung cancer to the pleura. We found that the MPM had R282W, a key TP53 mutation, and genome-wide allelic loss or loss of heterozygosity, a distinct genomic alteration not previously described in MPM. We identified frequent inactivating SETDB1 mutations in this patient and in 68 additional MPM patients (mutation frequency: 10%, 7/69) by targeted deep sequencing. Our observations suggest the possibility of a new genetic mechanism in the development of either MPM or multiple primary cancers. The frequent SETDB1 inactivating mutations suggest there could be new diagnostic or therapeutic options for MPM.
Collapse
Affiliation(s)
- Hio Chung Kang
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.,Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Hong Kwan Kim
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.,Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | - Pedro Mendez
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Gavitt Woodard
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jun-Hee Yoon
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Kuang-Yu Jen
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Li Tai Fang
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Kirk Jones
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - David M Jablons
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.,Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Il-Jin Kim
- Thoracic Oncology Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.,Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
24
|
Nabavi N, Bennewith KL, Churg A, Wang Y, Collins CC, Mutti L. Switching off malignant mesothelioma: exploiting the hypoxic microenvironment. Genes Cancer 2016; 7:340-354. [PMID: 28191281 PMCID: PMC5302036 DOI: 10.18632/genesandcancer.124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/31/2016] [Indexed: 12/21/2022] Open
Abstract
Malignant mesotheliomas are aggressive, asbestos-related cancers with poor patient prognosis, typically arising in the mesothelial surfaces of tissues in pleural and peritoneal cavity. The relative unspecific symptoms of mesotheliomas, misdiagnoses, and lack of precise targeted therapies call for a more critical assessment of this disease. In the present review, we categorize commonly identified genomic aberrations of mesotheliomas into their canonical pathways and discuss targeting these pathways in the context of tumor hypoxia, a hallmark of cancer known to render solid tumors more resistant to radiation and most chemo-therapy. We then explore the concept that the intrinsic hypoxic microenvironment of mesotheliomas can be Achilles' heel for targeted, multimodal therapeutic intervention.
Collapse
Affiliation(s)
- Noushin Nabavi
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, BC, Canada
- Department of Urologic Sciences, University of British Columbia, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, BC, Canada
| | - Kevin L. Bennewith
- Department of Integrative Oncology, BC Cancer Agency, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC, Canada
| | - Andrew Churg
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences, University of British Columbia, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, BC, Canada
| | - Colin C. Collins
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, BC, Canada
- Department of Urologic Sciences, University of British Columbia, BC, Canada
| | - Luciano Mutti
- Italian Group for Research and Therapy for Mesothelioma (GIMe) & School of Environment and Life Sciences, University of Salford, Manchester, United Kingdom
| |
Collapse
|
25
|
Sheffield BS, Fulton R, Kalloger SE, Milne K, Geller G, Jones M, Jacquemont C, Zachara S, Zhao E, Pleasance E, Laskin J, Jones SJM, Marra MA, Yip S, Nelson BH, Gown AM, Ho C, Ionescu DN. Investigation of PD-L1 Biomarker Testing Methods for PD-1 Axis Inhibition in Non-squamous Non-small Cell Lung Cancer. J Histochem Cytochem 2016; 64:587-600. [PMID: 27591097 DOI: 10.1369/0022155416665338] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/26/2016] [Indexed: 11/22/2022] Open
Abstract
Inhibitors of the programmed cell death 1 (PD-1) signaling axis have recently demonstrated efficacy and are rapidly being incorporated into the treatment of non-small cell lung cancers (NSCLCs). Despite clear benefits to certain patients, the association of these responses with a predictive biomarker remains uncertain. Several different biomarkers have been proposed, with differing results and conclusions. This study compares multiple methods of biomarker testing for treatment of NSCLCs with PD1-axis inhibitors. Tissue microarrays of matched primary and metastatic NSCLCs were used to compare four different PD-1 ligand (PD-L1) IHC techniques, as well as RNA ISH. Additional cases with whole genome and transcriptome data were assessed for molecular correlates of PD-L1 overexpression. Eighty cases were included in the IHC study. Multiple IHC methodologies showed a high rate of agreement (Kappa = 0.67). When calibrated to RNA expression, agreement improved significantly (Kappa = 0.90, p=0.0049). PD-L1 status of primary and metastatic tumors was discordant in 17 (22%) cases. This study suggests that different IHC methodologies for PD-L1 assessment provide slightly different results. There is significant discordance between the PD-L1 status of primary tumors and lymph node metastases. RNA ISH may be a useful adjunct to complement PD-L1 IHC testing.
Collapse
Affiliation(s)
- Brandon S Sheffield
- Department of Pathology, Abbotsford Regional Hospital and Cancer Centre, British
Columbia, Canada (BSS)
| | - Regan Fulton
- PhenoPath Laboratories, Seattle, Washington (RF, CJ, AMG)
| | - Steve E Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (BSS, SEK, SZ, SY)
| | - Katy Milne
- Trev and Joyce Deeley Research Centre BC Cancer ,Agency, Vancouver, British Columbia, Canada(KM, BHN)
| | - Georgia Geller
- BC Cancer Agency, Vancouver, British Columbia, CanadaDivision of Medical Oncology ,BC Cancer Agency, Vancouver, British Columbia, Canada(GG, JL, CH)
| | - Martin Jones
- BC Cancer Agency, Vancouver, British Columbia, CanadaCanada's Michael Smith Genome Sciences Centre ,BC Cancer Agency, Vancouver, British Columbia, Canada (MJ, EZ, EP, SJMJ, MAM)
| | | | - Susanna Zachara
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (BSS, SEK, SZ, SY)
| | - Eric Zhao
- BC Cancer Agency, Vancouver, British Columbia, CanadaCanada's Michael Smith Genome Sciences Centre ,BC Cancer Agency, Vancouver, British Columbia, Canada (MJ, EZ, EP, SJMJ, MAM)
| | - Erin Pleasance
- BC Cancer Agency, Vancouver, British Columbia, CanadaCanada's Michael Smith Genome Sciences Centre ,BC Cancer Agency, Vancouver, British Columbia, Canada (MJ, EZ, EP, SJMJ, MAM)
| | - Janessa Laskin
- BC Cancer Agency, Vancouver, British Columbia, CanadaDivision of Medical Oncology ,BC Cancer Agency, Vancouver, British Columbia, Canada(GG, JL, CH)
| | - Steven J M Jones
- BC Cancer Agency, Vancouver, British Columbia, CanadaCanada's Michael Smith Genome Sciences Centre ,BC Cancer Agency, Vancouver, British Columbia, Canada (MJ, EZ, EP, SJMJ, MAM)
| | - Marco A Marra
- BC Cancer Agency, Vancouver, British Columbia, CanadaCanada's Michael Smith Genome Sciences Centre ,BC Cancer Agency, Vancouver, British Columbia, Canada (MJ, EZ, EP, SJMJ, MAM)
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (BSS, SEK, SZ, SY)
| | - Brad H Nelson
- Trev and Joyce Deeley Research Centre BC Cancer ,Agency, Vancouver, British Columbia, Canada(KM, BHN)
| | - Allen M Gown
- PhenoPath Laboratories, Seattle, Washington (RF, CJ, AMG)
| | - Cheryl Ho
- BC Cancer Agency, Vancouver, British Columbia, CanadaDivision of Medical Oncology ,BC Cancer Agency, Vancouver, British Columbia, Canada(GG, JL, CH)
| | - Diana N Ionescu
- Department of Laboratory Medicine and Pathology , BC Cancer Agency, Vancouver, British Columbia, Canada(DNI)
| |
Collapse
|
26
|
Brevet M. Comparative genetics of diffuse malignant mesothelioma tumors of the peritoneumand pleura, with focus on BAP1 expression. Pleura Peritoneum 2016; 1:91-97. [PMID: 30911612 DOI: 10.1515/pp-2016-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022] Open
Abstract
Malignant mesothelioma (MM) is a malignancy arising from the mesothelial cells lining the thoracic and abdominal serosal cavities. The pleural space is the most commonly affected site, accounting for about 80% of cases, while peritoneum makes up the majority of the remaining 20%. The different types of mesotheliomas are generally considered as distinct diseases with specific risk factors, therapeutic strategies and prognoses. Epidemiological and clinical differences between pleural and peritoneal MM raise questions about the involvement of different molecular mechanisms. Since the BAP1 gene is involved in the BAP1 cancer syndrome and seems to be a prognostic factor in MM, this review presents an overview of BAP1 alterations in mesothelioma comparing pleural and peritoneal localizations.
Collapse
|
27
|
Lai J, Zhou Z, Tang XJ, Gao ZB, Zhou J, Chen SQ. A Tumor-Specific Neo-Antigen Caused by a Frameshift Mutation in BAP1 Is a Potential Personalized Biomarker in Malignant Peritoneal Mesothelioma. Int J Mol Sci 2016; 17:ijms17050739. [PMID: 27187383 PMCID: PMC4881561 DOI: 10.3390/ijms17050739] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/28/2016] [Accepted: 05/04/2016] [Indexed: 01/28/2023] Open
Abstract
Malignant peritoneal mesothelioma (MPM) is an aggressive rare malignancy associated with asbestos exposure. A better understanding of the molecular pathogenesis of MPM will help develop a targeted therapy strategy. Oncogene targeted depth sequencing was performed on a tumor sample and paired peripheral blood DNA from a patient with malignant mesothelioma of the peritoneum. Four somatic base-substitutions in NOTCH2, NSD1, PDE4DIP, and ATP10B and 1 insert frameshift mutation in BAP1 were validated by the Sanger method at the transcriptional level. A 13-amino acids neo-peptide of the truncated Bap1 protein, which was produced as a result of this novel frameshift mutation, was predicted to be presented by this patient's HLA-B protein. The polyclonal antibody of the synthesized 13-mer neo-peptide was produced in rabbits. Western blotting results showed a good antibody-neoantigen specificity, and Immunohistochemistry (IHC) staining with the antibody of the neo-peptide clearly differentiated neoplastic cells from normal cells. A search of the Catalogue of Somatic Mutations in Cancer (COSMIC) database also revealed that 53.2% of mutations in BAP1 were frameshift indels with neo-peptide formation. An identified tumor-specific neo-antigen could be the potential molecular biomarker for personalized diagnosis to precisely subtype rare malignancies such as MPM.
Collapse
Affiliation(s)
- Jun Lai
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Zhan Zhou
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Xiao-Jing Tang
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Zhi-Bin Gao
- Department of Obstetrics and Gynecology, Yuyao People's Hospital, 800 Chengdong Road, Yuyao 315400, China.
| | - Jie Zhou
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Shu-Qing Chen
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| |
Collapse
|
28
|
Sheffield BS, Lorette J, Shen Y, Marra MA, Churg A. Immunohistochemistry for NF2, LATS1/2, and YAP/TAZ Fails to Separate Benign From Malignant Mesothelial Proliferations. Arch Pathol Lab Med 2016; 140:391. [DOI: 10.5858/arpa.2015-0508-le] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | - Yaoqing Shen
- Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada
| | - Marco A. Marra
- Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada
- Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Andrew Churg
- Departments of Pathology and Laboratory Medicine and
- Department of Pathology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
29
|
Utility of BAP1 Immunohistochemistry and p16 (CDKN2A) FISH in the Diagnosis of Malignant Mesothelioma in Effusion Cytology Specimens. Am J Surg Pathol 2016; 40:120-6. [DOI: 10.1097/pas.0000000000000529] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Case BW. Pathology analysis for mesothelioma study in the United Kingdom: Current practice and historical development. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2016; 19:201-212. [PMID: 27705547 DOI: 10.1080/10937404.2016.1195320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Following up on the largest case-control study of malignant mesothelioma yet performed, investigators at the London School of Hygiene and Tropical Medicine assessed 1732 male and 670 female cases as of May 2013. Epidemiological findings of a subset of these were published previously, excluding patients who died or who refused to be interviewed. Pathology reports were collected for subjects, including those both eligible and ineligible for epidemiology study based on vital status. The current investigation examined 860 cases having pathology reports available. Sixty-one cases were diagnosed using cytology only, often with equivocal diagnoses, while 799 reported at least a biopsy of the tumor. Of these, 748 had pathology sufficiently detailed for evaluation. These reports were examined for basis of diagnosis, differences between study cases and ineligible cases, pathology characteristics, and immunohistochemical and other tests used. The most prominent subtype was epithelioid (64% of study cases but only 49% of ineligible cases). Biphasic subtype was present in 10% of study cases and 16% of those ineligible. Sarcomatoid subtype was present in 7% of study cases and 19% of ineligible cases, most of whom died. Twelve percent of study cases displayed no specified subtype, versus 7% of ineligible cases. Of recorded immunohistochemical stains specific for mesothelial cell origin, calretinin (95%) and CK 5/6 or CK5 alone (84%) were by far the most common. Calretinin and CK 5/6 or CK 5 alone were also most sensitive and positive in 92% of cases presenting with surgical pathology report. Ninety percent of cases had at least one immunohistochemical marker for possible lung carcinoma applied, with BER-Ep4 and TTF-1 the most frequent at 68% and CEA at 58%. TTF-1 and CEA were positive in 1% or less of cases. Patterns of use and positive and negative results for each of these as well as other immunohistochemical stains are presented and discussed, along with a brief historical description of their development and use. Possible effects of the pathologic analysis on the results of previously published and future epidemiological studies are discussed.
Collapse
Affiliation(s)
- B W Case
- a Department of Pathology and School of Environment , McGill University , Montreal , Québec , Canada
| |
Collapse
|
31
|
Claimon A, Bang JI, Cheon GJ, Kim EE, Lee DS. Malignant Peritoneal Mesothelioma Masquerades as Peritoneal Metastasis on (18)F-FDG PET/CT Scans; a Rare Diagnosis that Should Not Be Missed. Nucl Med Mol Imaging 2015; 49:325-8. [PMID: 26550054 DOI: 10.1007/s13139-015-0360-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 07/05/2015] [Accepted: 07/30/2015] [Indexed: 10/23/2022] Open
Abstract
Malignant peritoneal mesothelioma (MPM) is a rare but fatal tumor. The clinical presentations and imaging findings are nonspecific and resemble various diseases, including peritoneal metastasis. Imaging findings of MPH on (18)F-(18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography (PET)/computed tomography (CT) are diverse and not well described. We report the two cases of biopsy-proven MPH using (18)F-FDG PET/CT. In our cases, interesting disease patterns-including MPH arising from visceral peritoneal lining of kidney that suffer from polycystic disease and from the parietal peritoneum beneath the appendectomy scar-were presented. One case showed classical metastases localized within the abdominal cavity; while the other case exhibited the rare pattern of extensive multi-organ metastases. By knowing the possible variations and diagnostic pitfalls of (18)F-FDG PET/CT findings in MPM, more accurate interpretation of such mysterious cancer is attainable.
Collapse
Affiliation(s)
- Apichaya Claimon
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehangno, Jongro-gu Seoul, 110-744 Korea ; Division of Nuclear Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ji-In Bang
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehangno, Jongro-gu Seoul, 110-744 Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehangno, Jongro-gu Seoul, 110-744 Korea
| | - Euishin Edmund Kim
- Molecular Medicine and Biopharmaceutical Sciences, WCU Graduate School of Convergence Science and Technology, Seoul National University College of Medicine, Seoul, Korea ; Department of Radiological Sciences, University of California, Irvine, CA USA
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehangno, Jongro-gu Seoul, 110-744 Korea ; Molecular Medicine and Biopharmaceutical Sciences, WCU Graduate School of Convergence Science and Technology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Next-Generation Sequencing Approaches in Cancer: Where Have They Brought Us and Where Will They Take Us? Cancers (Basel) 2015; 7:1925-58. [PMID: 26404381 PMCID: PMC4586802 DOI: 10.3390/cancers7030869] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022] Open
Abstract
Next-generation sequencing (NGS) technologies and data have revolutionized cancer research and are increasingly being deployed to guide clinicians in treatment decision-making. NGS technologies have allowed us to take an “omics” approach to cancer in order to reveal genomic, transcriptomic, and epigenomic landscapes of individual malignancies. Integrative multi-platform analyses are increasingly used in large-scale projects that aim to fully characterize individual tumours as well as general cancer types and subtypes. In this review, we examine how NGS technologies in particular have contributed to “omics” approaches in cancer research, allowing for large-scale integrative analyses that consider hundreds of tumour samples. These types of studies have provided us with an unprecedented wealth of information, providing the background knowledge needed to make small-scale (including “N of 1”) studies informative and relevant. We also take a look at emerging opportunities provided by NGS and state-of-the-art third-generation sequencing technologies, particularly in the context of translational research. Cancer research and care are currently poised to experience significant progress catalyzed by accessible sequencing technologies that will benefit both clinical- and research-based efforts.
Collapse
|
33
|
Bose P, Pleasance ED, Jones M, Shen Y, Ch’ng C, Reisle C, Schein JE, Mungall AJ, Moore R, Ma Y, Sheffield BS, Thomson T, Rasmussen S, Ng T, Yip S, Lee CW, Ho C, Laskin J, Marra MA, Jones SJ. Integrative genomic analysis of ghost cell odontogenic carcinoma. Oral Oncol 2015; 51:e71-5. [DOI: 10.1016/j.oraloncology.2015.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 12/14/2022]
|