1
|
Hamdan A, Ewing A. Unravelling the tumour genome: The evolutionary and clinical impacts of structural variants in tumourigenesis. J Pathol 2022; 257:479-493. [PMID: 35355264 PMCID: PMC9321913 DOI: 10.1002/path.5901] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 11/15/2022]
Abstract
Structural variants (SVs) represent a major source of aberration in tumour genomes. Given the diversity in the size and type of SVs present in tumours, the accurate detection and interpretation of SVs in tumours is challenging. New classes of complex structural events in tumours are discovered frequently, and the definitions of the genomic consequences of complex events are constantly being refined. Detailed analyses of short-read whole-genome sequencing (WGS) data from large tumour cohorts facilitate the interrogation of SVs at orders of magnitude greater scale and depth. However, the inherent technical limitations of short-read WGS prevent us from accurately detecting and investigating the impact of all the SVs present in tumours. The expanded use of long-read WGS will be critical for improving the accuracy of SV detection, and in fully resolving complex SV events, both of which are crucial for determining the impact of SVs on tumour progression and clinical outcome. Despite the present limitations, we demonstrate that SVs play an important role in tumourigenesis. In particular, SVs contribute significantly to late-stage tumour development and to intratumoural heterogeneity. The evolutionary trajectories of SVs represent a window into the clonal dynamics in tumours, a comprehensive understanding of which will be vital for influencing patient outcomes in the future. Recent findings have highlighted many clinical applications of SVs in cancer, from early detection to biomarkers for treatment response and prognosis. As the methods to detect and interpret SVs improve, elucidating the full breadth of the complex SV landscape and determining how these events modulate tumour evolution will improve our understanding of cancer biology and our ability to capitalise on the utility of SVs in the clinical management of cancer patients. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Alhafidz Hamdan
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Cancer Research UK Edinburgh Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Ailith Ewing
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Cancer Research UK Edinburgh Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
2
|
Harber J, Kamata T, Pritchard C, Fennell D. Matter of TIME: the tumor-immune microenvironment of mesothelioma and implications for checkpoint blockade efficacy. J Immunother Cancer 2021; 9:e003032. [PMID: 34518291 PMCID: PMC8438820 DOI: 10.1136/jitc-2021-003032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an incurable cancer with a dismal prognosis and few effective treatment options. Nonetheless, recent positive phase III trial results for immune checkpoint blockade (ICB) in MPM herald a new dawn in the fight to advance effective treatments for this cancer. Tumor mutation burden (TMB) has been widely reported to predict ICB in other cancers, but MPM is considered a low-TMB tumor. Similarly, tumor programmed death-ligand 1 (PD-L1) expression has not been proven predictive in phase III clinical trials in MPM. Consequently, the precise mechanisms that determine response to immunotherapy in this cancer remain unknown. The present review therefore aimed to synthesize our current understanding of the tumor immune microenvironment in MPM and reflects on how specific cellular features might impact immunotherapy responses or lead to resistance. This approach will inform stratified approaches to therapy and advance immunotherapy combinations in MPM to improve clinical outcomes further.
Collapse
Affiliation(s)
- James Harber
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Tamihiro Kamata
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Catrin Pritchard
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Dean Fennell
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| |
Collapse
|
3
|
Hotta K, Fujimoto N. Current evidence and future perspectives of immune-checkpoint inhibitors in unresectable malignant pleural mesothelioma. J Immunother Cancer 2021; 8:jitc-2019-000461. [PMID: 32098830 PMCID: PMC7057421 DOI: 10.1136/jitc-2019-000461] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Platinum-based chemotherapy is commonly used as the standard first-line treatment for unresectable malignant pleural mesothelioma (MPM). However, in recent times, immune-checkpoint inhibitors (ICIs) have led to a paradigm shift. Herein, we review relevant literature and ongoing trials of ICIs used as both first-line and salvage therapies. Specifically, in the Japanese single-arm, phase II trial, the MERIT trial, nivolumab, an antiprogrammed cell death 1 (PD-1) antibody showed favorable efficacy when used as a salvage therapy. Currently, multiple ICI monotherapy or combination therapy trials have been conducted, which could provide further evidence. Among available ICIs, the anti-PD-1 antibody is promising for unresectable MPM, despite the limited efficacy of anti-CTLA4 monotherapy. Ongoing studies will further confirm the potential efficacy of ICIs for MPM, as observed across other malignancies. It is also crucial to identify any clinically useful predictive biomarkers that could reveal ICIs with maximal effects in MPM.
Collapse
Affiliation(s)
- Katsuyuki Hotta
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Nobukazu Fujimoto
- Department of Medical Oncology and Medicine, Okayama Rosai Hospitalosai Hospital, Okayama, Japan
| |
Collapse
|
4
|
Hiltbrunner S, Mannarino L, Kirschner MB, Opitz I, Rigutto A, Laure A, Lia M, Nozza P, Maconi A, Marchini S, D’Incalci M, Curioni-Fontecedro A, Grosso F. Tumor Immune Microenvironment and Genetic Alterations in Mesothelioma. Front Oncol 2021; 11:660039. [PMID: 34249695 PMCID: PMC8261295 DOI: 10.3389/fonc.2021.660039] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare and fatal disease of the pleural lining. Up to 80% of the MPM cases are linked to asbestos exposure. Even though its use has been banned in the industrialized countries, the cases continue to increase. MPM is a lethal cancer, with very little survival improvements in the last years, mirroring very limited therapeutic advances. Platinum-based chemotherapy in combination with pemetrexed and surgery are the standard of care, but prognosis is still unacceptably poor with median overall survival of approximately 12 months. The genomic landscape of MPM has been widely characterized showing a low mutational burden and the impairment of tumor suppressor genes. Among them, BAP1 and BLM are present as a germline inactivation in a small subset of patients and increases predisposition to tumorigenesis. Other studies have demonstrated a high frequency of mutations in DNA repair genes. Many therapy approaches targeting these alterations have emerged and are under evaluation in the clinic. High-throughput technologies have allowed the detection of more complex molecular events, like chromotripsis and revealed different transcriptional programs for each histological subtype. Transcriptional analysis has also paved the way to the study of tumor-infiltrating cells, thus shedding lights on the crosstalk between tumor cells and the microenvironment. The tumor microenvironment of MPM is indeed crucial for the pathogenesis and outcome of this disease; it is characterized by an inflammatory response to asbestos exposure, involving a variety of chemokines and suppressive immune cells such as M2-like macrophages and regulatory T cells. Another important feature of MPM is the dysregulation of microRNA expression, being frequently linked to cancer development and drug resistance. This review will give a detailed overview of all the above mentioned features of MPM in order to improve the understanding of this disease and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Stefanie Hiltbrunner
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, University of Zurich, Zurich, Switzerland
| | - Laura Mannarino
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | | | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Angelica Rigutto
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, University of Zurich, Zurich, Switzerland
| | - Alexander Laure
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, University of Zurich, Zurich, Switzerland
| | - Michela Lia
- Mesothelioma Unit, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Paolo Nozza
- Department of Pathology, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Antonio Maconi
- Infrastruttura Ricerca Formazione Innovazione (IRFI), Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Sergio Marchini
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - Maurizio D’Incalci
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - Alessandra Curioni-Fontecedro
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, University of Zurich, Zurich, Switzerland
| | - Federica Grosso
- Mesothelioma Unit, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
- Translational Medicine, Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| |
Collapse
|
5
|
Menis J, Pasello G, Remon J. Immunotherapy in malignant pleural mesothelioma: a review of literature data. Transl Lung Cancer Res 2021; 10:2988-3000. [PMID: 34295692 PMCID: PMC8264322 DOI: 10.21037/tlcr-20-673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/04/2021] [Indexed: 01/09/2023]
Abstract
Malignant pleural mesothelioma (MPM) is a rare, aggressive cancer of the pleural surface, associated with asbestos exposure, whose incidence is still growing in some areas of the world. MPM is still considered a rare and an orphan disease with an unchanged median overall survival (OS) ranging from 8 to 14 months and no treatment advances in the last 15 years both in local and advanced disease. In the recent years, chronic inflammation of the mesothelium together with local tumor suppression plays a major role in the malignant transformation. Also, significant heterogeneity in both tumor and the microenvironment is at the basis of MPM biology. Preclinical data have demonstrated the immunogenicity and the lack of an effective antitumor response by the immune system in MPM thus paving the way to the development of immune therapeutics in this disease. Still there is no clear evidence of any predictive biomarker so that, given the close interaction between the immune infiltrate and mesothelial cells, a number of trials are ongoing to investigate the role and prognostic value of the immune microenvironment. In this review we summarize the rationale for immune therapeutics development in MPM, as well as, the relevant literature and ongoing trials of immune checkpoint inhibitors (ICIs) and vaccines used as both first-line treatment and beyond.
Collapse
Affiliation(s)
- Jessica Menis
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy;,Medical Oncology Department, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Giulia Pasello
- Medical Oncology Department, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Jordi Remon
- Department of Medical Oncology, Centro Integral Oncológico Clara Campal (HM-CIOCC), Hospital HM Delfos, HM Hospitales, Barcelona, Spain
| |
Collapse
|
6
|
Carbone M, Adusumilli PS, Alexander HR, Baas P, Bardelli F, Bononi A, Bueno R, Felley-Bosco E, Galateau-Salle F, Jablons D, Mansfield AS, Minaai M, de Perrot M, Pesavento P, Rusch V, Severson DT, Taioli E, Tsao A, Woodard G, Yang H, Zauderer MG, Pass HI. Mesothelioma: Scientific clues for prevention, diagnosis, and therapy. CA Cancer J Clin 2019; 69:402-429. [PMID: 31283845 PMCID: PMC8192079 DOI: 10.3322/caac.21572] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesothelioma affects mostly older individuals who have been occupationally exposed to asbestos. The global mesothelioma incidence and mortality rates are unknown, because data are not available from developing countries that continue to use large amounts of asbestos. The incidence rate of mesothelioma has decreased in Australia, the United States, and Western Europe, where the use of asbestos was banned or strictly regulated in the 1970s and 1980s, demonstrating the value of these preventive measures. However, in these same countries, the overall number of deaths from mesothelioma has not decreased as the size of the population and the percentage of old people have increased. Moreover, hotspots of mesothelioma may occur when carcinogenic fibers that are present in the environment are disturbed as rural areas are being developed. Novel immunohistochemical and molecular markers have improved the accuracy of diagnosis; however, about 14% (high-resource countries) to 50% (developing countries) of mesothelioma diagnoses are incorrect, resulting in inadequate treatment and complicating epidemiological studies. The discovery that germline BRCA1-asssociated protein 1 (BAP1) mutations cause mesothelioma and other cancers (BAP1 cancer syndrome) elucidated some of the key pathogenic mechanisms, and treatments targeting these molecular mechanisms and/or modulating the immune response are being tested. The role of surgery in pleural mesothelioma is controversial as it is difficult to predict who will benefit from aggressive management, even when local therapies are added to existing or novel systemic treatments. Treatment outcomes are improving, however, for peritoneal mesothelioma. Multidisciplinary international collaboration will be necessary to improve prevention, early detection, and treatment.
Collapse
Affiliation(s)
- Michele Carbone
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Prasad S. Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - H. Richard Alexander
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Paul Baas
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Fabrizio Bardelli
- National Research Council Institute of Nanotechnology, La Sapienza University, Rome, Italy
| | - Angela Bononi
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Raphael Bueno
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Division of Thoracic Surgery, University Hospital of Zurich, Zurich, Switzerland
| | | | - David Jablons
- Thoracic Oncology, Department of Surgery, Helen Diller Cancer Center, University of California at San Francisco, San Francisco, California
| | | | - Michael Minaai
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Marc de Perrot
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Patricia Pesavento
- Pathology, Immunology, and Microbiology Laboratory, University of California at Davis, Sacramento, California
| | - Valerie Rusch
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David T. Severson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emanuela Taioli
- Translational Epidemiology and Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anne Tsao
- Division of Cancer Medicine, Department of Thoracic and Head/Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gavitt Woodard
- Thoracic Oncology, Department of Surgery, Helen Diller Cancer Center, University of California at San Francisco, San Francisco, California
| | - Haining Yang
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | | | - Harvey I. Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York
| |
Collapse
|
7
|
Nambirajan A, Malgulwar PB, Sharma A, Boorgula MT, Doddamani R, Singh M, Suri V, Sarkar C, Sharma MC. Clinicopathological evaluation of PD-L1 expression and cytotoxic T-lymphocyte infiltrates across intracranial molecular subgroups of ependymomas: are these tumors potential candidates for immune check-point blockade? Brain Tumor Pathol 2019; 36:152-161. [PMID: 31388782 DOI: 10.1007/s10014-019-00350-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/25/2019] [Indexed: 02/02/2023]
Abstract
Immune check-point blockade (ICB) targeting programmed cell death ligand-1 (PD-L1)/programmed death-1 (PD-1) axis has created paradigm shift in cancer treatment. 'ST-RELA' and 'PF-A' molecular subgroups of ependymomas (EPN) show poor outcomes. We aimed to understand the potential candidature of EPNs for ICB. Supratentorial (ST) Grade II/III EPNs were classified into ST-RELA, ST-YAP, and ST-not otherwise specified (NOS), based on RELA/YAP1 fusion transcripts and/or L1CAM and p65 protein expression. Posterior fossa (PF) EPNs were classified into PF-A and PF-B based on H3K27me3 expression. Immunohistochemistry for PD-L1 and CD8 was performed. RelA protein enrichment at PDL1 promoter site was analysed by chromatin immunoprecipitation-qPCR (ChIP-qPCR). Eighty-three intracranial EPNs were studied. Median tumor infiltrating CD8 + cytotoxic T-lymphocyte (CTL) density was 6/mm2, and was higher in ST-EPNs (median 10/mm2) as compared to PF-EPNs (median 3/mm2). PD-L1 expression was noted in 17/83 (20%) EPNs, including 12/31 ST-RELA and rare ST-NOS (2/12), PF-A (2/25) and PF-B (1/13) EPNs. Twelve EPNs (14%) showed high CTL density and concurrent PD-L1 positivity, of which majority (10/12) were ST-RELA EPNs. Enrichment of RelA protein was seen at PDL1 promoter. Increased CTL densities and upregulation of PD-L1 in ST-RELA ependymomas suggests potential candidature for immunotherapy.
Collapse
Affiliation(s)
- Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Prit Benny Malgulwar
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Agrima Sharma
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Meher Thej Boorgula
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ramesh Doddamani
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Manmohan Singh
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Mehar Chand Sharma
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|