1
|
Kadariya Y, Sementino E, Hua X, Kappes DJ, Testa JR. Modeling Malignant Mesothelioma in Genetically Engineered Mice. Curr Protoc 2025; 5:e70086. [PMID: 39791266 PMCID: PMC11737608 DOI: 10.1002/cpz1.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Mesothelioma is a lethal cancer of the serosal lining of the body cavities. Risk factors include environmental and genetic factors. Asbestos exposure is considered the principal environmental risk factor, but other carcinogenic mineral fibers, such as erionite, also have a causal role. Pathogenic germline (heritable) mutations of specific genes, especially BAP1, are thought to predispose the individual to mesothelioma in about 10% of cases. Somatic mutations and deletions of specific tumor suppressor genes, particularly BAP1, CDKN2A/B, and NF2, occur frequently in human mesothelioma, and asbestos-exposed mice with heterozygous deletions of any one of these genes have been shown to develop mesothelioma more often and at an accelerated rate than in control animals. Autochthonous mesothelioma mouse models, which are genetically engineered to carry multiple genetic lesions matching those observed in the human disease counterpart, closely resemble the disease phenotype and the extensive inflammatory responses that characterize human mesothelioma. Because autochthonous mice do not require asbestos exposure and form tumors rapidly, these models are invaluable for assessing novel therapeutic strategies in an immunocompetent setting. The overlapping genetic, epigenetic, and immune environments of the tumors observed in these genetically engineered mouse models (GEMMs) and human primary mesothelioma specimens support the clinical relevance of these preclinical models. This article presents protocols for studies of asbestos-induced mesothelioma in GEMMs and non-carcinogenic conditional knockout models of mesothelioma, including an example of a preclinical application. These models are invaluable for understanding the biological underpinnings of mesothelioma and for testing new therapeutics and chemoprevention or interception agents. © 2025 Wiley Periodicals LLC. Basic Protocol 1: Generation of a genetically engineered mouse model (GEMM) with a germline Bap1 knockout allele Basic Protocol 2: Generation of GEMMs with germline Bap1 knock-in alleles Basic Protocol 3: Asbestos carcinogenicity investigations with GEMMs Basic Protocol 4: Preclinical chemoprevention and chemotherapy studies using a GEMM with asbestos-induced mesothelioma Basic Protocol 5: Generation of a GEMM with conditional knockout of Bap1 Basic Protocol 6: Generation of a conditional knockout model of mesothelioma.
Collapse
Affiliation(s)
- Yuwaraj Kadariya
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Eleonora Sementino
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Xiang Hua
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Dietmar J. Kappes
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Joseph R. Testa
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Zhu R, Liu X, Zhang X, Zhong Z, Qi S, Jin R, Gu Y, Wang Y, Ling C, Chen K, Ye D, Yu FX. Gene therapy for diffuse pleural mesotheliomas in preclinical models by concurrent expression of NF2 and SuperHippo. Cell Rep Med 2024; 5:101763. [PMID: 39368484 PMCID: PMC11513813 DOI: 10.1016/j.xcrm.2024.101763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/03/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Diffuse pleural mesothelioma (DPM) is a lethal cancer with a poor prognosis and limited treatment options. The Hippo signaling pathway genes, such as NF2 and LATS1/2, are frequently mutated in DPM, indicating a tumor suppressor role in the development of DPM. Here, we show that in DPM cell lines lacking NF2 and in mice with a conditional Nf2 knockout, downregulation of WWC proteins, another family of Hippo pathway regulators, accelerates DPM progression. Conversely, the expression of SuperHippo, a WWC-derived minigene, effectively enhances Hippo signaling and suppresses DPM development. Moreover, the adeno-associated virus serotype 6 (AAV6) has been engineered to deliver both NF2 and SuperHippo genes into mesothelial cells, which substantially impedes tumor growth in xenograft and genetic DPM models and prolongs the median survival of mice. These findings serve as a proof of concept for the potential use of gene therapy targeting the Hippo pathway to treat DPM.
Collapse
Affiliation(s)
- Rui Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xincheng Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sixian Qi
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ruxin Jin
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuan Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dan Ye
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Xu D, Yin S, Shu Y. NF2: An underestimated player in cancer metabolic reprogramming and tumor immunity. NPJ Precis Oncol 2024; 8:133. [PMID: 38879686 PMCID: PMC11180135 DOI: 10.1038/s41698-024-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024] Open
Abstract
Neurofibromatosis type 2 (NF2) is a tumor suppressor gene implicated in various tumors, including mesothelioma, schwannomas, and meningioma. As a member of the ezrin, radixin, and moesin (ERM) family of proteins, merlin, which is encoded by NF2, regulates diverse cellular events and signalling pathways, such as the Hippo, mTOR, RAS, and cGAS-STING pathways. However, the biological role of NF2 in tumorigenesis has not been fully elucidated. Furthermore, cross-cancer mutations may exert distinct biological effects on tumorigenesis and treatment response. In addition to the functional inactivation of NF2, the codeficiency of other genes, such as cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B), BRCA1-associated protein-1 (BAP1), and large tumor suppressor 2 (LATS2), results in unique tumor characteristics that should be considered in clinical treatment decisions. Notably, several recent studies have explored the metabolic and immunological features associated with NF2, offering potential insights into tumor biology and the development of innovative therapeutic strategies. In this review, we consolidate the current knowledge on NF2 and examine the potential connection between cancer metabolism and tumor immunity in merlin-deficient malignancies. This review may provide a deeper understanding of the biological roles of NF2 and guide possible therapeutic avenues.
Collapse
Affiliation(s)
- Duo Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyuan Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Hillen H, Candi A, Vanderhoydonck B, Kowalczyk W, Sansores-Garcia L, Kesikiadou EC, Van Huffel L, Spiessens L, Nijs M, Soons E, Haeck W, Klaassen H, Smets W, Spieser SA, Marchand A, Chaltin P, Ciesielski F, Debaene F, Chen L, Kamal A, Gwaltney SL, Versele M, Halder GA. A Novel Irreversible TEAD Inhibitor, SWTX-143, Blocks Hippo Pathway Transcriptional Output and Causes Tumor Regression in Preclinical Mesothelioma Models. Mol Cancer Ther 2024; 23:3-13. [PMID: 37748190 DOI: 10.1158/1535-7163.mct-22-0681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/13/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
The Hippo pathway and its downstream effectors, the YAP and TAZ transcriptional coactivators, are deregulated in multiple different types of human cancer and are required for cancer cell phenotypes in vitro and in vivo, while largely dispensable for tissue homeostasis in adult mice. YAP/TAZ and their main partner transcription factors, the TEAD1-4 factors, are therefore promising anticancer targets. Because of frequent YAP/TAZ hyperactivation caused by mutations in the Hippo pathway components NF2 and LATS2, mesothelioma is one of the prime cancer types predicted to be responsive to YAP/TAZ-TEAD inhibitor treatment. Mesothelioma is a devastating disease for which currently no effective treatment options exist. Here, we describe a novel covalent YAP/TAZ-TEAD inhibitor, SWTX-143, that binds to the palmitoylation pocket of all four TEAD isoforms. SWTX-143 caused irreversible and specific inhibition of the transcriptional activity of YAP/TAZ-TEAD in Hippo-mutant tumor cell lines. More importantly, YAP/TAZ-TEAD inhibitor treatment caused strong mesothelioma regression in subcutaneous xenograft models with human cells and in an orthotopic mesothelioma mouse model. Finally, SWTX-143 also selectively impaired the growth of NF2-mutant kidney cancer cell lines, suggesting that the sensitivity of mesothelioma models to these YAP/TAZ-TEAD inhibitors can be extended to other tumor types with aberrations in Hippo signaling. In brief, we describe a novel and specific YAP/TAZ-TEAD inhibitor that has potential to treat multiple Hippo-mutant solid tumor types.
Collapse
Affiliation(s)
- Hanne Hillen
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | | | | | - Weronika Kowalczyk
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leticia Sansores-Garcia
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Elena C Kesikiadou
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leen Van Huffel
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Lore Spiessens
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | - Patrick Chaltin
- Cistim Leuven vzw, Leuven, Belgium
- Center for Drug Design and Discovery (CD3), KU Leuven, Leuven, Belgium
| | | | | | - Lei Chen
- SpringWorks Therapeutics, Stamford, Connecticut
| | | | | | | | - Georg A Halder
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Bertuccio FR, Agustoni F, Galli G, Bortolotto C, Saddi J, Baietto G, Baio N, Montini S, Putignano P, D’Ambrosio G, Corsico AG, Pedrazzoli P, Stella GM. Pleural Mesothelioma: Treatable Traits of a Heterogeneous Disease. Cancers (Basel) 2023; 15:5731. [PMID: 38136277 PMCID: PMC10741585 DOI: 10.3390/cancers15245731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/23/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Pleural mesothelioma is an aggressive disease with diffuse nature, low median survival, and prolonged latency presenting difficulty in prognosis, diagnosis, and treatment. Here, we review all these aspects to underline the progress being made in its investigation and to emphasize how much work remains to be carried out to improve prognosis and treatment.
Collapse
Affiliation(s)
- Francesco Rocco Bertuccio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Francesco Agustoni
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Department of Medical Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulia Galli
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Department of Medical Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Jessica Saddi
- Department of Oncology, Clinical-Surgical, Unit of Radiation Therapy, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Guido Baietto
- Cardiothoracic and Vascular Department, Unit of Thoracic Surgery, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Nicola Baio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Simone Montini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paola Putignano
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Gioacchino D’Ambrosio
- Pathology Unit, Department of Diagnostical Services and Imaging, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Angelo G. Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paolo Pedrazzoli
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Department of Medical Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (F.R.B.); (F.A.); (G.G.); (N.B.); (S.M.); (P.P.); (A.G.C.); (P.P.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
6
|
Farahmand P, Gyuraszova K, Rooney C, Raffo-Iraolagoitia XL, Jayasekera G, Hedley A, Johnson E, Chernova T, Malviya G, Hall H, Monteverde T, Blyth K, Duffin R, Carlin LM, Lewis D, Le Quesne J, MacFarlane M, Murphy DJ. Asbestos accelerates disease onset in a genetic model of malignant pleural mesothelioma. FRONTIERS IN TOXICOLOGY 2023; 5:1200650. [PMID: 37441092 PMCID: PMC10333928 DOI: 10.3389/ftox.2023.1200650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Hypothesis: Asbestos-driven inflammation contributes to malignant pleural mesothelioma beyond the acquisition of rate-limiting mutations. Methods: Genetically modified conditional allelic mice that were previously shown to develop mesothelioma in the absence of exposure to asbestos were induced with lentiviral vector expressing Cre recombinase with and without intrapleural injection of amosite asbestos and monitored until symptoms required euthanasia. Resulting tumours were examined histologically and by immunohistochemistry for expression of lineage markers and immune cell infiltration. Results: Injection of asbestos dramatically accelerated disease onset and end-stage tumour burden. Tumours developed in the presence of asbestos showed increased macrophage infiltration. Pharmacological suppression of macrophages in mice with established tumours failed to extend survival or to enhance response to chemotherapy. Conclusion: Asbestos-driven inflammation contributes to the severity of mesothelioma beyond the acquisition of rate-limiting mutations, however, targeted suppression of macrophages in established epithelioid mesothelioma showed no therapeutic benefit.
Collapse
Affiliation(s)
- Pooyeh Farahmand
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Claire Rooney
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Respiratory Medicine, Royal Infirmary, Glasgow, United Kingdom
| | | | - Geeshath Jayasekera
- Glasgow Pleural Disease Unit, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Ann Hedley
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Emma Johnson
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Tatyana Chernova
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Gaurav Malviya
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Holly Hall
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Tiziana Monteverde
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kevin Blyth
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Glasgow Pleural Disease Unit, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Rodger Duffin
- Centre for Inflammation Research, Edinburgh, United Kingdom
| | - Leo M. Carlin
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - David Lewis
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - John Le Quesne
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Department of Histopathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Marion MacFarlane
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Daniel J. Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
7
|
Sato Y, Elbadawy M, Suzuki K, Tsunedomi R, Nagano H, Ishihara Y, Yamamoto H, Azakami D, Uchide T, Nabeta R, Fukushima R, Abugomaa A, Kaneda M, Yamawaki H, Shinohara Y, Usui T, Sasaki K. Establishment of an experimental model of canine malignant mesothelioma organoid culture using a three-dimensional culture method. Biomed Pharmacother 2023; 162:114651. [PMID: 37030135 DOI: 10.1016/j.biopha.2023.114651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
Canine malignant mesothelioma (cMM) is a rare and drug-resistant malignant tumor. Due to few patients and experimental models, there have not been enough studies to demonstrate the pathogenesis of the disease and novel effective treatment for cMM. Since cMM resembles human MM (hMM) in histopathological characteristics, it is also considered a promising research model of hMM. Compared with conventional 2-dimensional (2D) culture methods, 3-dimensional (3D) organoid culture can recapitulate the properties of original tumor tissues. However, cMM organoids have never been developed. In the present study, we for the first time generated cMM organoids using the pleural effusion samples. Organoids from individual MM dogs were successfully generated. They exhibited the characteristics of MM and expressed mesothelial cell markers, such as WT-1 and mesothelin. The sensitivity to anti-cancer drugs was different in each strain of cMM organoids. RNA sequencing analysis showed cell adhesion molecule pathways were specifically upregulated in cMM organoids compared with their corresponding 2D cultured cells. Among these genes, the expression level of E-cadherin was drastically higher in the organoids than that in the 2D cells. In conclusion, our established cMM organoids might become a new experimental tool to provide new insights into canine and human MM therapy.
Collapse
Affiliation(s)
- Yomogi Sato
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, 13736, Moshtohor, Toukh, Elqaliobiya, Egypt.
| | - Kazuhiko Suzuki
- Laboratory of Veterinary Toxicology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yusuke Ishihara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Haru Yamamoto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Daigo Azakami
- Laboratory of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Tsuyoshi Uchide
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Rina Nabeta
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Ryuji Fukushima
- Animal Medical Emergency Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; Faculty of Veterinary Medicine, Mansoura University, 35516 Mansoura, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1, Higashi 23 ban-cho, Towada, Aomori 034-8628, Japan
| | - Yuta Shinohara
- Pet Health & Food Division, Iskara Industry CO., LTD, 1-14-2, Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan.
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| |
Collapse
|
8
|
Piccolo S, Panciera T, Contessotto P, Cordenonsi M. YAP/TAZ as master regulators in cancer: modulation, function and therapeutic approaches. NATURE CANCER 2023; 4:9-26. [PMID: 36564601 PMCID: PMC7614914 DOI: 10.1038/s43018-022-00473-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
Our understanding of the function of the transcriptional regulators YAP and TAZ (YAP/TAZ) in cancer is advancing. In this Review, we provide an update on recent progress in YAP/TAZ biology, their regulation by Hippo signaling and mechanotransduction and highlight open questions. YAP/TAZ signaling is an addiction shared by multiple tumor types and their microenvironments, providing many malignant attributes. As such, it represents an important vulnerability that may offer a broad window of therapeutic efficacy, and here we give an overview of the current treatment strategies and pioneering clinical trials.
Collapse
Affiliation(s)
- Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- IFOM-ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Tito Panciera
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | | |
Collapse
|
9
|
Hiltbrunner S, Fleischmann Z, Sokol ES, Zoche M, Felley-Bosco E, Curioni-Fontecedro A. Genomic landscape of pleural and peritoneal mesothelioma tumours. Br J Cancer 2022; 127:1997-2005. [PMID: 36138075 PMCID: PMC9681755 DOI: 10.1038/s41416-022-01979-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Malignant pleural and peritoneal mesotheliomas are rare malignancies with unacceptable poor prognoses and limited treatment options. The genomic landscape is mainly characterised by the loss of tumour suppressor genes and mutations in DNA repair genes. Currently, data from next-generation sequencing (NGS) of mesothelioma tumours is restricted to a limited number of cases; moreover, data comparing molecular features of mesothelioma from the pleural and peritoneal origin with NGS are lacking. METHODS We analysed 1113 pleural mesothelioma and 355 peritoneal mesothelioma samples. All tumours were sequenced with the FoundationOne® or FoundationOne®CDx assay for detection of substitutions, insertion-deletions, copy-number alterations and selected rearrangements in at least 324 cancer genes. RESULTS This analysis revealed alterations in 19 genes with an overall prevalence of at least 2%. Alterations in BAP1, CDKN2A, CDKN2B, NF2, MTAP, TP53 and SETD2 occurred with a prevalence of at least 10%. Peritoneal, compared to pleural mesothelioma, was characterised by a lower prevalence of alterations in CDKN2A, CDKN2B and MTAP. Moreover, we could define four distinct subgroups according to alterations in BAP1 and CDKN2A/B. Alterations in Hedgehog pathway-related genes (PTCH1/2 and SUFU) and Hippo pathway-related gene (NF2) as well as KRAS, EGFR, PDGFRA/B, ERBB2 and FGFR3 were detected in both cohorts. CONCLUSION Here, we report the molecular aberrations from the largest cohort of patients with mesothelioma. This analysis identified a proportion of patients with targetable alterations and suggests that molecular profiling can identify new treatment options for patients with mesothelioma.
Collapse
Affiliation(s)
- Stefanie Hiltbrunner
- grid.412004.30000 0004 0478 9977Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland ,grid.7400.30000 0004 1937 0650University of Zurich, Zurich, Switzerland
| | - Zoe Fleischmann
- grid.418158.10000 0004 0534 4718Foundation Medicine, Cambridge, MA USA
| | - Ethan S. Sokol
- grid.418158.10000 0004 0534 4718Foundation Medicine, Cambridge, MA USA
| | - Martin Zoche
- grid.412004.30000 0004 0478 9977Pathology Department, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Emanuela Felley-Bosco
- grid.7400.30000 0004 1937 0650University of Zurich, Zurich, Switzerland ,grid.412004.30000 0004 0478 9977Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Alessandra Curioni-Fontecedro
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland. .,University of Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Gao Y, Kruithof-de Julio M, Peng RW, Dorn P. Organoids as a Model for Precision Medicine in Malignant Pleural Mesothelioma: Where Are We Today? Cancers (Basel) 2022; 14:3758. [PMID: 35954422 PMCID: PMC9367391 DOI: 10.3390/cancers14153758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022] Open
Abstract
MPM is an aggressive tumor originating from pleural mesothelial cells. A characteristic feature of the disease is the dominant prevalence of therapeutically intractable inactivating alterations in TSGs, making MPM one of the most difficult cancers to treat and the epitome of a cancer characterized by a significant lack of therapy options and an extremely poor prognosis (5-year survival rate of only 5% to 10%). Extensive interpatient heterogeneity poses another major challenge for targeted therapy of MPM, warranting stratified therapy for specific subgroups of MPM patients. Accurate preclinical models are critical for the discovery of new therapies and the development of personalized medicine. Organoids, an in vitro 'organ-like' 3D structure derived from patient tumor tissue that faithfully mimics the biology and complex architecture of cancer and largely overcomes the limitations of other existing models, are the next-generation tumor model. Although organoids have been successfully produced and used in many cancers, the development of MPM organoids is still in its infancy. Here, we provide an overview of recent advances in cancer organoids, focusing on the progress and challenges in MPM organoid development. We also elaborate the potential of MPM organoids for understanding MPM pathobiology, discovering new therapeutic targets, and developing personalized treatments for MPM patients.
Collapse
Affiliation(s)
- Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland;
- Department of BioMedical Research (DBMR), Oncology-Thoracic Malignancies (OTM), University of Bern, 3008 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland;
- Department for BioMedical Research (DBMR), Translation Organoid Research, University of Bern, 3008 Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, 3008 Bern, Switzerland
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland;
- Department of BioMedical Research (DBMR), Oncology-Thoracic Malignancies (OTM), University of Bern, 3008 Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland;
- Department of BioMedical Research (DBMR), Oncology-Thoracic Malignancies (OTM), University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
11
|
Song Y, Baxter SS, Dai L, Sanders C, Burkett S, Baugher RN, Mellott SD, Young TB, Lawhorn HE, Difilippantonio S, Karim B, Kadariya Y, Pinto LA, Testa JR, Shoemaker RH. Mesothelioma Mouse Models with Mixed Genomic States of Chromosome and Microsatellite Instability. Cancers (Basel) 2022; 14:3108. [PMID: 35804881 PMCID: PMC9264972 DOI: 10.3390/cancers14133108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Malignant mesothelioma (MMe) is a rare malignancy originating from the linings of the pleural, peritoneal and pericardial cavities. The best-defined risk factor is exposure to carcinogenic mineral fibers (e.g., asbestos). Genomic studies have revealed that the most frequent genetic lesions in human MMe are mutations in tumor suppressor genes. Several genetically engineered mouse models have been generated by introducing the same genetic lesions found in human MMe. However, most of these models require specialized breeding facilities and long-term exposure of mice to asbestos for MMe development. Thus, an alternative model with high tumor penetrance without asbestos is urgently needed. We characterized an orthotopic model using MMe cells derived from Cdkn2a+/-;Nf2+/- mice chronically injected with asbestos. These MMe cells were tumorigenic upon intraperitoneal injection. Moreover, MMe cells showed mixed chromosome and microsatellite instability, supporting the notion that genomic instability is relevant in MMe pathogenesis. In addition, microsatellite markers were detectable in the plasma of tumor-bearing mice, indicating a potential use for early cancer detection and monitoring the effects of interventions. This orthotopic model with rapid development of MMe without asbestos exposure represents genomic instability and specific molecular targets for therapeutic or preventive interventions to enable preclinical proof of concept for the intervention in an immunocompetent setting.
Collapse
Affiliation(s)
- Yurong Song
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Shaneen S. Baxter
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Lisheng Dai
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Chelsea Sanders
- Animal Research Technical Support of Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (C.S.); (S.D.)
| | - Sandra Burkett
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD 21702, USA;
| | - Ryan N. Baugher
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Stephanie D. Mellott
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Todd B. Young
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Heidi E. Lawhorn
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Simone Difilippantonio
- Animal Research Technical Support of Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (C.S.); (S.D.)
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Yuwaraj Kadariya
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (Y.K.); (J.R.T.)
| | - Ligia A. Pinto
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Joseph R. Testa
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (Y.K.); (J.R.T.)
| | - Robert H. Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20892, USA;
| |
Collapse
|
12
|
Kuryk L, Rodella G, Staniszewska M, Pancer KW, Wieczorek M, Salmaso S, Caliceti P, Garofalo M. Novel Insights Into Mesothelioma Therapy: Emerging Avenues and Future Prospects. Front Oncol 2022; 12:916839. [PMID: 35785199 PMCID: PMC9247278 DOI: 10.3389/fonc.2022.916839] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022] Open
Abstract
Malignant mesothelioma is a rare and aggressive cancer that develops in the thin layer surrounding the mesothelium and is mainly caused by asbestos exposure. Despite improvements in patient prognosis with conventional cancer treatments, such as surgery, chemotherapy, and radiotherapy, there are still no curative treatment modalities for advanced disease. In recent years, new therapeutic avenues have been explored. Improved understanding of the mechanisms underlying the dynamic tumor interaction with the immune system has led to the development of immunotherapeutic approaches. Numerous recent clinical trials have shown a desire to develop more effective treatments that can be used to fight against the disease. Immune checkpoint inhibitors, oncolytic adenoviruses, and their combination represent a promising strategy that can be used to synergistically overcome immunosuppression in the mesothelioma tumor microenvironment. This review provides a synthesized overview of the current state of knowledge on new therapeutic options for mesothelioma with a focus on the results of clinical trials conducted in the field.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health National Institute of Hygiene (NIH)—National Institute of Research, Warsaw, Poland
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Giulia Rodella
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Katarzyna Wanda Pancer
- Department of Virology, National Institute of Public Health National Institute of Hygiene (NIH)—National Institute of Research, Warsaw, Poland
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health National Institute of Hygiene (NIH)—National Institute of Research, Warsaw, Poland
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Sementino E, Kadariya Y, Cheung M, Menges CW, Tan Y, Kukuyan AM, Shrestha U, Karchugina S, Cai KQ, Peri S, Duncan JS, Chernoff J, Testa JR. Inactivation of p21-Activated Kinase 2 (Pak2) Inhibits the Development of Nf2-Deficient Tumors by Restricting Downstream Hedgehog and Wnt Signaling. Mol Cancer Res 2022; 20:699-711. [PMID: 35082167 PMCID: PMC9081258 DOI: 10.1158/1541-7786.mcr-21-0837] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/02/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022]
Abstract
Because loss of the NF2 tumor suppressor gene results in p21-activated kinase (Pak) activation, PAK inhibitors hold promise for the treatment of NF2-deficient tumors. To test this possibility, we asked if loss of Pak2, a highly expressed group I PAK member, affects the development of malignant mesothelioma in Nf2;Cdkn2a-deficient (NC) mice and the growth properties of NC mesothelioma cells in culture. In vivo, deletion of Pak2 resulted in a markedly decreased incidence and delayed onset of both pleural and peritoneal malignant mesotheliomas in NC mice. In vitro, Pak2 deletion decreased malignant mesothelioma cell viability, migration, clonogenicity, and spheroid formation. RNA-sequencing analysis demonstrated downregulated expression of Hedgehog and Wnt pathway genes in NC;Pak2-/- mesothelioma cells versus NC;Pak2+/+ mesothelioma cells. Targeting of the Hedgehog signaling component Gli1 or its target gene Myc inhibited cell viability and spheroid formation in NC;P+/+ mesothelioma cells. Kinome profiling uncovered kinase changes indicative of EMT in NC;Pak2-/- mesothelioma cells, suggesting that Pak2-deficient malignant mesotheliomas can adapt by reprogramming their kinome in the absence of Pak activity. The identification of such compensatory pathways offers opportunities for rational combination therapies to circumvent resistance to anti-PAK drugs. IMPLICATIONS We provide evidence supporting a role for PAK inhibitors in treating NF2-deficient tumors. NF2-deficient tumors lacking Pak2 eventually adapt by kinome reprogramming, presenting opportunities for combination therapies to bypass anti-PAK drug resistance.
Collapse
Affiliation(s)
- Eleonora Sementino
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yuwaraj Kadariya
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mitchell Cheung
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Craig W. Menges
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yinfei Tan
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Anna-Mariya Kukuyan
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ujjawal Shrestha
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sofiia Karchugina
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kathy Q. Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Suraj Peri
- Bioinformatics and Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - James S. Duncan
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Jonathan Chernoff
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Joseph R. Testa
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Joseph R. Testa, Ph.D., Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 1911; Phone: (215) 728-2610; Fax: (215) 214-1619;
| |
Collapse
|
14
|
Shah R, Klotz LV, Glade J. Current Management and Future Perspective in Pleural Mesothelioma. Cancers (Basel) 2022; 14:1044. [PMID: 35205798 PMCID: PMC8869935 DOI: 10.3390/cancers14041044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/16/2022] Open
Abstract
Pleural mesothelioma is an aggressive malignancy arising from pleural mesothelial cell lining, predominantly associated with prior exposure to asbestos. The ban on asbestos use has led to its lower incidence in many countries, but globally the disease burden is expected to rise. Therefore, well-planned research is needed to develop more effective, tolerable and affordable drugs. The development of novel treatment has been too slow, with only two regimens of systemic therapy with robust phase 3 data approved formally to date. The treatment scenario for resectable disease remains controversial. However, recent developments in the understanding of disease and clinical trials have been encouraging, and may add better treatment options in the coming years. In this review, we discuss the current treatment options for pleural mesothelioma and shed light on some recent studies and ongoing trials.
Collapse
Affiliation(s)
- Rajiv Shah
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Laura V. Klotz
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University Hospital, 69126 Heidelberg, Germany;
| | - Julia Glade
- Institute for Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| |
Collapse
|
15
|
Marazioti A, Krontira AC, Behrend SJ, Giotopoulou GA, Ntaliarda G, Blanquart C, Bayram H, Iliopoulou M, Vreka M, Trassl L, Pepe MAA, Hackl CM, Klotz LV, Weiss SAI, Koch I, Lindner M, Hatz RA, Behr J, Wagner DE, Papadaki H, Antimisiaris SG, Jean D, Deshayes S, Grégoire M, Kayalar Ö, Mortazavi D, Dilege Ş, Tanju S, Erus S, Yavuz Ö, Bulutay P, Fırat P, Psallidas I, Spella M, Giopanou I, Lilis I, Lamort A, Stathopoulos GT. KRAS signaling in malignant pleural mesothelioma. EMBO Mol Med 2022; 14:e13631. [PMID: 34898002 PMCID: PMC8819314 DOI: 10.15252/emmm.202013631] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 10/28/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) arises from mesothelial cells lining the pleural cavity of asbestos-exposed individuals and rapidly leads to death. MPM harbors loss-of-function mutations in BAP1, NF2, CDKN2A, and TP53, but isolated deletion of these genes alone in mice does not cause MPM and mouse models of the disease are sparse. Here, we show that a proportion of human MPM harbor point mutations, copy number alterations, and overexpression of KRAS with or without TP53 changes. These are likely pathogenic, since ectopic expression of mutant KRASG12D in the pleural mesothelium of conditional mice causes epithelioid MPM and cooperates with TP53 deletion to drive a more aggressive disease form with biphasic features and pleural effusions. Murine MPM cell lines derived from these tumors carry the initiating KRASG12D lesions, secondary Bap1 alterations, and human MPM-like gene expression profiles. Moreover, they are transplantable and actionable by KRAS inhibition. Our results indicate that KRAS alterations alone or in accomplice with TP53 alterations likely play an important and underestimated role in a proportion of patients with MPM, which warrants further exploration.
Collapse
|
16
|
Abemaciclib in patients with p16ink4A-deficient mesothelioma (MiST2): a single-arm, open-label, phase 2 trial. Lancet Oncol 2022; 23:374-381. [DOI: 10.1016/s1470-2045(22)00062-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/23/2022]
|
17
|
Okazaki Y. Asbestos‐induced mesothelial injury and carcinogenesis: Involvement of iron and reactive oxygen species. Pathol Int 2021; 72:83-95. [DOI: 10.1111/pin.13196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/11/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Yasumasa Okazaki
- Department of Pathology and Biological Responses Nagoya University Graduate School of Medicine Showa‐Ku Nagoya Japan
| |
Collapse
|
18
|
Shamseddin M, Obacz J, Garnett MJ, Rintoul RC, Francies HE, Marciniak SJ. Use of preclinical models for malignant pleural mesothelioma. Thorax 2021; 76:1154-1162. [PMID: 33692175 PMCID: PMC8526879 DOI: 10.1136/thoraxjnl-2020-216602] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/16/2021] [Accepted: 02/26/2021] [Indexed: 01/08/2023]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer most commonly caused by prior exposure to asbestos. Median survival is 12-18 months, since surgery is ineffective and chemotherapy offers minimal benefit. Preclinical models that faithfully recapitulate the genomic and histopathological features of cancer are critical for the development of new treatments. The most commonly used models of MPM are two-dimensional cell lines established from primary tumours or pleural fluid. While these have provided some important insights into MPM biology, these cell models have significant limitations. In order to address some of these limitations, spheroids and microfluidic chips have more recently been used to investigate the role of the three-dimensional environment in MPM. Efforts have also been made to develop animal models of MPM, including asbestos-induced murine tumour models, MPM-prone genetically modified mice and patient-derived xenografts. Here, we discuss the available in vitro and in vivo models of MPM and highlight their strengths and limitations. We discuss how newer technologies, such as the tumour-derived organoids, might allow us to address the limitations of existing models and aid in the identification of effective treatments for this challenging-to-treat disease.
Collapse
Affiliation(s)
- Marie Shamseddin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Joanna Obacz
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Mathew J Garnett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Robert Campbell Rintoul
- Department of Oncology, University of Cambridge, Cambridge, Cambridgeshire, UK
- Department of Thoracic Oncology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | | | - Stefan John Marciniak
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, Cambridgeshire, UK
| |
Collapse
|
19
|
Obacz J, Yung H, Shamseddin M, Linnane E, Liu X, Azad AA, Rassl DM, Fairen-Jimenez D, Rintoul RC, Nikolić MZ, Marciniak SJ. Biological basis for novel mesothelioma therapies. Br J Cancer 2021; 125:1039-1055. [PMID: 34226685 PMCID: PMC8505556 DOI: 10.1038/s41416-021-01462-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/13/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Mesothelioma is an aggressive cancer that is associated with exposure to asbestos. Although asbestos is banned in several countries, including the UK, an epidemic of mesothelioma is predicted to affect middle-income countries during this century owing to their heavy consumption of asbestos. The prognosis for patients with mesothelioma is poor, reflecting a failure of conventional chemotherapy that has ultimately resulted from an inadequate understanding of its biology. However, recent work has revolutionised the study of mesothelioma, identifying genetic and pathophysiological vulnerabilities, including the loss of tumour suppressors, epigenetic dysregulation and susceptibility to nutrient stress. We discuss how this knowledge, combined with advances in immunotherapy, is enabling the development of novel targeted therapies.
Collapse
Affiliation(s)
- Joanna Obacz
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Henry Yung
- UCL Respiratory, Division of Medicine Rayne Institute, University College London, London, UK
| | - Marie Shamseddin
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Saffron Walden, UK
| | - Emily Linnane
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Xiewen Liu
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Arsalan A Azad
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Doris M Rassl
- Department of Histopathology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - David Fairen-Jimenez
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Robert C Rintoul
- Department of Oncology, University of Cambridge, Cambridge, UK
- Department of Thoracic Oncology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Marko Z Nikolić
- UCL Respiratory, Division of Medicine Rayne Institute, University College London, London, UK
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
20
|
Grosso S, Marini A, Gyuraszova K, Voorde JV, Sfakianos A, Garland GD, Tenor AR, Mordue R, Chernova T, Morone N, Sereno M, Smith CP, Officer L, Farahmand P, Rooney C, Sumpton D, Das M, Teodósio A, Ficken C, Martin MG, Spriggs RV, Sun XM, Bushell M, Sansom OJ, Murphy D, MacFarlane M, Le Quesne JPC, Willis AE. The pathogenesis of mesothelioma is driven by a dysregulated translatome. Nat Commun 2021; 12:4920. [PMID: 34389715 PMCID: PMC8363647 DOI: 10.1038/s41467-021-25173-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 07/25/2021] [Indexed: 12/22/2022] Open
Abstract
Malignant mesothelioma (MpM) is an aggressive, invariably fatal tumour that is causally linked with asbestos exposure. The disease primarily results from loss of tumour suppressor gene function and there are no 'druggable' driver oncogenes associated with MpM. To identify opportunities for management of this disease we have carried out polysome profiling to define the MpM translatome. We show that in MpM there is a selective increase in the translation of mRNAs encoding proteins required for ribosome assembly and mitochondrial biogenesis. This results in an enhanced rate of mRNA translation, abnormal mitochondrial morphology and oxygen consumption, and a reprogramming of metabolic outputs. These alterations delimit the cellular capacity for protein biosynthesis, accelerate growth and drive disease progression. Importantly, we show that inhibition of mRNA translation, particularly through combined pharmacological targeting of mTORC1 and 2, reverses these changes and inhibits malignant cell growth in vitro and in ex-vivo tumour tissue from patients with end-stage disease. Critically, we show that these pharmacological interventions prolong survival in animal models of asbestos-induced mesothelioma, providing the basis for a targeted, viable therapeutic option for patients with this incurable disease.
Collapse
Affiliation(s)
- Stefano Grosso
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Alberto Marini
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Katarina Gyuraszova
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | | | | | - Gavin D Garland
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Angela Rubio Tenor
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Ryan Mordue
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Tanya Chernova
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Nobu Morone
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Marco Sereno
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Claire P Smith
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Leah Officer
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Pooyeh Farahmand
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Claire Rooney
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - David Sumpton
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Madhumita Das
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Ana Teodósio
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Catherine Ficken
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Maria Guerra Martin
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Ruth V Spriggs
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Xiao-Ming Sun
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Owen J Sansom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Daniel Murphy
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - Marion MacFarlane
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK.
| | - John P C Le Quesne
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK.
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK.
- Glenfield Hospital, Groby Road, University Hospitals Leicester NHS Trust Leicester, Leicester, UK.
| | - Anne E Willis
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK.
| |
Collapse
|
21
|
Behrouzfar K, Burton K, Mutsaers SE, Morahan G, Lake RA, Fisher SA. How to Better Understand the Influence of Host Genetics on Developing an Effective Immune Response to Thoracic Cancers. Front Oncol 2021; 11:679609. [PMID: 34235080 PMCID: PMC8256168 DOI: 10.3389/fonc.2021.679609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/31/2021] [Indexed: 01/02/2023] Open
Abstract
Thoracic cancers pose a significant global health burden. Immune checkpoint blockade therapies have improved treatment outcomes, but durable responses remain limited. Understanding how the host immune system interacts with a developing tumor is essential for the rational development of improved treatments for thoracic malignancies. Recent technical advances have improved our understanding of the mutational burden of cancer cells and changes in cancer-specific gene expression, providing a detailed understanding of the complex biology underpinning tumor-host interactions. While there has been much focus on the genetic alterations associated with cancer cells and how they may impact treatment outcomes, how host genetics affects cancer development is also critical and will greatly determine treatment response. Genome-wide association studies (GWAS) have identified genetic variants associated with cancer predisposition. This approach has successfully identified host genetic risk factors associated with common thoracic cancers like lung cancer, but is less effective for rare cancers like malignant mesothelioma. To assess how host genetics impacts rare thoracic cancers, we used the Collaborative Cross (CC); a powerful murine genetic resource designed to maximize genetic diversity and rapidly identify genes associated with any biological trait. We are using the CC in conjunction with our asbestos-induced MexTAg mouse model, to identify host genes associated with mesothelioma development. Once genes that moderate tumor development and progression are known, human homologues can be identified and human datasets interrogated to validate their association with disease outcome. Furthermore, our CC-MexTAg animal model enables in-depth study of the tumor microenvironment, allowing the correlation of immune cell infiltration and gene expression signatures with disease development. This strategy provides a detailed picture of the underlying biological pathways associated with mesothelioma susceptibility and progression; knowledge that is crucial for the rational development of new diagnostic and therapeutic strategies. Here we discuss the influence of host genetics on developing an effective immune response to thoracic cancers. We highlight current knowledge gaps, and with a focus on mesothelioma, describe the development and application of the CC-MexTAg to overcome limitations and illustrate how the knowledge gained from this unique study will inform the rational design of future treatments of mesothelioma.
Collapse
Affiliation(s)
- Kiarash Behrouzfar
- National Centre for Asbestos Related Diseases (NCARD), University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Kimberley Burton
- National Centre for Asbestos Related Diseases (NCARD), University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Steve E. Mutsaers
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
- Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases (NCARD), University of Western Australia, Nedlands, WA, Australia
| | - Scott A. Fisher
- National Centre for Asbestos Related Diseases (NCARD), University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
22
|
Vita E, Stefani A, Di Salvatore M, Chiappetta M, Lococo F, Margaritora S, Tortora G, Bria E. Oncological Frontiers in the Treatment of Malignant Pleural Mesothelioma. J Clin Med 2021; 10:2290. [PMID: 34070352 PMCID: PMC8197557 DOI: 10.3390/jcm10112290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/09/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy characterized by very poor prognosis and lack of treatment options. Immunotherapy has rapidly emerged as an effective tool for MPM, particularly for tumors of non-epithelioid histology. At the same time, comprehensive genomic sequencing may open the way to new-generation targeted-drugs able to hit specific MPM molecular vulnerabilities. These innovations will possibly enrich, but also dramatically complicate, the elucidation of treatment algorithms. Multidisciplinary integration is urgently needed.
Collapse
Affiliation(s)
- Emanuele Vita
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (A.S.); (M.D.S.); (G.T.)
- Medical Oncology, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Alessio Stefani
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (A.S.); (M.D.S.); (G.T.)
- Medical Oncology, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Mariantonietta Di Salvatore
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (A.S.); (M.D.S.); (G.T.)
- Medical Oncology, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Marco Chiappetta
- Thoracy Surgery, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.C.); (F.L.); (S.M.)
| | - Filippo Lococo
- Thoracy Surgery, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.C.); (F.L.); (S.M.)
| | - Stefano Margaritora
- Thoracy Surgery, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.C.); (F.L.); (S.M.)
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (A.S.); (M.D.S.); (G.T.)
- Medical Oncology, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (A.S.); (M.D.S.); (G.T.)
- Medical Oncology, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
23
|
Zhang M, Luo JL, Sun Q, Harber J, Dawson AG, Nakas A, Busacca S, Sharkey AJ, Waller D, Sheaff MT, Richards C, Wells-Jordan P, Gaba A, Poile C, Baitei EY, Bzura A, Dzialo J, Jama M, Le Quesne J, Bajaj A, Martinson L, Shaw JA, Pritchard C, Kamata T, Kuse N, Brannan L, De Philip Zhang P, Yang H, Griffiths G, Wilson G, Swanton C, Dudbridge F, Hollox EJ, Fennell DA. Clonal architecture in mesothelioma is prognostic and shapes the tumour microenvironment. Nat Commun 2021; 12:1751. [PMID: 33741915 PMCID: PMC7979861 DOI: 10.1038/s41467-021-21798-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/04/2021] [Indexed: 01/31/2023] Open
Abstract
Malignant Pleural Mesothelioma (MPM) is typically diagnosed 20-50 years after exposure to asbestos and evolves along an unknown evolutionary trajectory. To elucidate this path, we conducted multi-regional exome sequencing of 90 tumour samples from 22 MPMs acquired at surgery. Here we show that exomic intratumour heterogeneity varies widely across the cohort. Phylogenetic tree topology ranges from linear to highly branched, reflecting a steep gradient of genomic instability. Using transfer learning, we detect repeated evolution, resolving 5 clusters that are prognostic, with temporally ordered clonal drivers. BAP1/-3p21 and FBXW7/-chr4 events are always early clonal. In contrast, NF2/-22q events, leading to Hippo pathway inactivation are predominantly late clonal, positively selected, and when subclonal, exhibit parallel evolution indicating an evolutionary constraint. Very late somatic alteration of NF2/22q occurred in one patient 12 years after surgery. Clonal architecture and evolutionary clusters dictate MPM inflammation and immune evasion. These results reveal potentially drugable evolutionary bottlenecking in MPM, and an impact of clonal architecture on shaping the immune landscape, with potential to dictate the clinical response to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Min Zhang
- Novogene Co., Ltd, Building 301, Beijing, China
| | - Jin-Li Luo
- Bioinformatics and Biostatistics Support Hub, University of Leicester, Leicester, UK
| | | | - James Harber
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Alan G Dawson
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
- Department of Cardiothoracic Surgery, Glenfield Hospital, Leicester, UK
| | - Apostolos Nakas
- Department of Cardiothoracic Surgery, Glenfield Hospital, Leicester, UK
| | - Sara Busacca
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | | | - David Waller
- Barts Health NHS Trust, The Royal London Hospital, London, UK
| | | | - Cathy Richards
- Department of Pathology, Leicester Royal Infirmary, Infirmary Square, Leicester, Leicestershire, UK
| | - Peter Wells-Jordan
- Department of Pathology, Leicester Royal Infirmary, Infirmary Square, Leicester, Leicestershire, UK
| | - Aarti Gaba
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Charlotte Poile
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Essa Y Baitei
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Aleksandra Bzura
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Joanna Dzialo
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Maymun Jama
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - John Le Quesne
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Bearsden, UK
| | - Amrita Bajaj
- Department of Radiology, Glenfield Hospital, Leicester, UK
| | - Luke Martinson
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Jacqui A Shaw
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Catrin Pritchard
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Tamihiro Kamata
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Nathaniel Kuse
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Lee Brannan
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | - Hongji Yang
- Department of Informatics, University of Leicester, Leicester, UK
| | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | | | | | - Frank Dudbridge
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Dean A Fennell
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
24
|
Badhai J, Pandey GK, Song JY, Krijgsman O, Bhaskaran R, Chandrasekaran G, Kwon MC, Bombardelli L, Monkhorst K, Grasso C, Zevenhoven J, van der Vliet J, Cozijnsen M, Krimpenfort P, Peeper D, van Lohuizen M, Berns A. Combined deletion of Bap1, Nf2, and Cdkn2ab causes rapid onset of malignant mesothelioma in mice. J Exp Med 2021; 217:151644. [PMID: 32271879 PMCID: PMC7971132 DOI: 10.1084/jem.20191257] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/09/2019] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
We have generated mouse models of malignant mesothelioma (MM) based upon disruption of the Bap1, Nf2, and Cdkn2ab tumor suppressor loci in various combinations as also frequently observed in human MM. Inactivation of all three loci in the mesothelial lining of the thoracic cavity leads to a highly aggressive MM that recapitulates the histological features and gene expression profile observed in human patients. The tumors also show a similar inflammatory phenotype. Bap1 deletion alone does not cause MM but dramatically accelerates MM development when combined with Nf2 and Cdkn2ab (hereafter BNC) disruption. The accelerated tumor development is accompanied by increased Polycomb repression and EZH2-mediated redistribution of H3K27me3 toward promoter sites with concomitant activation of PI3K and MAPK pathways. Treatment of BNC tumor–bearing mice with cisplatin and pemetrexed, the current frontline treatment, prolongs survival. This makes the autochthonous mouse model described here very well suited to explore the pathogenesis of MM and validate new treatment regimens for MM, including immunotherapy.
Collapse
Affiliation(s)
- Jitendra Badhai
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gaurav Kumar Pandey
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Oscar Krijgsman
- Oncode Institute, Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Rajith Bhaskaran
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gayathri Chandrasekaran
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Min-Chul Kwon
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Lorenzo Bombardelli
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Kim Monkhorst
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Cristoforo Grasso
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - John Zevenhoven
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jan van der Vliet
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Miranda Cozijnsen
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Paul Krimpenfort
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Daniel Peeper
- Oncode Institute, Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maarten van Lohuizen
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Anton Berns
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
25
|
Asciak R, George V, Rahman NM. Update on biology and management of mesothelioma. Eur Respir Rev 2021; 30:30/159/200226. [PMID: 33472960 DOI: 10.1183/16000617.0226-2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022] Open
Abstract
Malignant pleural mesothelioma is an aggressive, incurable cancer that is usually caused by asbestos exposure several decades before symptoms arise. Despite widespread prohibition of asbestos production and supply, its incidence continues to increase. It is heterogeneous in its presentation and behaviour, and diagnosis can be notoriously difficult. Identification of actionable gene mutations has proven challenging and current treatment options are largely ineffective, with a median survival of 10-12 months.However, the past few years have witnessed major advances in our understanding of the biology and pathogenesis of mesothelioma. This has also revealed the limitations of existing diagnostic algorithms and identified new treatment targets.Recent clinical trials have re-examined the role of surgery, provided new options for the management of associated pleural effusions and heralded the addition of targeted therapies. The increasing complexity of mesothelioma management, along with a desperate need for further research, means that a multidisciplinary team framework is essential for the delivery of contemporary mesothelioma care.This review provides a synthesised overview of the current state of knowledge and an update on the latest research in the field.
Collapse
Affiliation(s)
- Rachelle Asciak
- Oxford Centre for Respiratory Medicine, University Hospitals NHS Foundation Trust, Oxford, UK .,Mater Dei Hospital, Msida, Malta
| | - Vineeth George
- Oxford Centre for Respiratory Medicine, University Hospitals NHS Foundation Trust, Oxford, UK
| | - Najiib M Rahman
- Oxford Centre for Respiratory Medicine, University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
26
|
Seastedt KP, Pruett N, Hoang CD. Mouse models for mesothelioma drug discovery and development. Expert Opin Drug Discov 2020; 16:697-708. [PMID: 33380218 DOI: 10.1080/17460441.2021.1867530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Mesothelioma is an aggressive mesothelial lining tumor. Available drug therapies include chemotherapeutic agents, targeted molecular therapies, and immune system modulators. Mouse models were instrumental in the discovery and evaluation of such therapies, but there is need for improved understanding of the role of inflammation, tumor heterogeneity, mechanisms of carcinogenesis, and the tumor microenvironment. Novel mouse models may provide new insights and drive drug therapy discovery that improves efficacy. AREAS COVERED This review concerns available mouse models for mesothelioma drug discovery and development including the advantages and disadvantages of each. Gaps in current knowledge of mesothelioma are highlighted, and future directions for mouse model research are considered. EXPERT OPINION Soon, CRISPR-Cas gene-editing will improve understanding of mesothelioma mechanisms foundational to the discovery and testing of efficacious therapeutic targets. There are at least two likely areas of upcoming methodology development. One is concerned with precise modeling of inflammation - is it a causal process whereby inflammatory signals contribute to tumor initiation, or is it a secondary passenger process driven by asbestos exposure effects? The other area of methods improvement regards the availability of humanized immunocompromised mice harboring patient-derived xenografts. Combining human tumors in an environment with human immune cells will enable rapid innovation in immuno-oncology therapeutics.
Collapse
Affiliation(s)
- Kenneth P Seastedt
- Department of Surgery, Uniformed Services University of the Health Sciences F. Edward Hébert School of Medicine, Bethesda, Maryland, USA
| | - Nathanael Pruett
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chuong D Hoang
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Xu D, Yang H, Schmid RA, Peng RW. Therapeutic Landscape of Malignant Pleural Mesothelioma: Collateral Vulnerabilities and Evolutionary Dependencies in the Spotlight. Front Oncol 2020; 10:579464. [PMID: 33072611 PMCID: PMC7538645 DOI: 10.3389/fonc.2020.579464] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is the epitome of a recalcitrant cancer driven by pharmacologically intractable tumor suppressor proteins. A significant but largely unmet challenge in the field is the translation of genetic information on alterations in tumor suppressor genes (TSGs) into effective cancer-specific therapies. The notion that abnormal tumor genome subverts physiological cellular processes, which creates collateral vulnerabilities contextually related to specific genetic alterations, offers a promising strategy to target TSG-driven MPM. Moreover, emerging evidence has increasingly appreciated the therapeutic potential of genetic and pharmacological dependencies acquired en route to cancer development and drug resistance. Here, we review the most recent progress on vulnerabilities co-selected by functional loss of major TSGs and dependencies evolving out of cancer development and resistance to cisplatin based chemotherapy, the only first-line regimen approved by the US Food and Drug Administration (FDA). Finally, we highlight CRISPR-based functional genomics that has emerged as a powerful platform for cancer drug discovery in MPM. The repertoire of MPM-specific “Achilles heel” rises on the horizon, which holds the promise to elucidate therapeutic landscape and may promote precision oncology for MPM.
Collapse
Affiliation(s)
- Duo Xu
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Haitang Yang
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralph A Schmid
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ren-Wang Peng
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Cakiroglu E, Senturk S. Genomics and Functional Genomics of Malignant Pleural Mesothelioma. Int J Mol Sci 2020; 21:ijms21176342. [PMID: 32882916 PMCID: PMC7504302 DOI: 10.3390/ijms21176342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare, aggressive cancer of the mesothelial cells lining the pleural surface of the chest wall and lung. The etiology of MPM is strongly associated with prior exposure to asbestos fibers, and the median survival rate of the diagnosed patients is approximately one year. Despite the latest advancements in surgical techniques and systemic therapies, currently available treatment modalities of MPM fail to provide long-term survival. The increasing incidence of MPM highlights the need for finding effective treatments. Targeted therapies offer personalized treatments in many cancers. However, targeted therapy in MPM is not recommended by clinical guidelines mainly because of poor target definition. A better understanding of the molecular and cellular mechanisms and the predictors of poor clinical outcomes of MPM is required to identify novel targets and develop precise and effective treatments. Recent advances in the genomics and functional genomics fields have provided groundbreaking insights into the genomic and molecular profiles of MPM and enabled the functional characterization of the genetic alterations. This review provides a comprehensive overview of the relevant literature and highlights the potential of state-of-the-art genomics and functional genomics research to facilitate the development of novel diagnostics and therapeutic modalities in MPM.
Collapse
Affiliation(s)
- Ece Cakiroglu
- Izmir Biomedicine and Genome Center, Izmir 35340, Turkey;
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir 35340, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir 35340, Turkey;
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir 35340, Turkey
- Correspondence:
| |
Collapse
|
29
|
Blanquart C, Jaurand MC, Jean D. The Biology of Malignant Mesothelioma and the Relevance of Preclinical Models. Front Oncol 2020; 10:388. [PMID: 32269966 PMCID: PMC7109283 DOI: 10.3389/fonc.2020.00388] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Malignant mesothelioma (MM), especially its more frequent form, malignant pleural mesothelioma (MPM), is a devastating thoracic cancer with limited therapeutic options. Recently, clinical trials that used immunotherapy strategies have yielded promising results, but the benefits are restricted to a limited number of patients. To develop new therapeutic strategies and define predictors of treatment response to existing therapy, better knowledge of the cellular and molecular mechanisms of MM tumors and sound preclinical models are needed. This review aims to provide an overview of our present knowledge and issues on both subjects. MM shows a complex pattern of molecular changes, including genetic, chromosomic, and epigenetic alterations. MM is also a heterogeneous cancer. The recently described molecular classifications for MPM could better consider inter-tumor heterogeneity, while histo-molecular gradients are an interesting way to consider both intra- and inter-tumor heterogeneities. Classical preclinical models are based on use of MM cell lines in culture or implanted in rodents, i.e., xenografts in immunosuppressed mice or isografts in syngeneic rodents to assess the anti-tumor immune response. Recent developments are tumoroids, patient-derived xenografts (PDX), xenografts in humanized mice, and genetically modified mice (GEM) that carry mutations identified in human MM tumor cells. Multicellular tumor spheroids are an interesting in vitro model to reduce animal experimentation; they are more accessible than tumoroids. They could be relevant, especially if they are co-cultured with stromal and immune cells to partially reproduce the human microenvironment. Even if preclinical models have allowed for major advances, they show several limitations: (i) the anatomical and biological tumor microenvironments are incompletely reproduced; (ii) the intra-tumor heterogeneity and immunological contexts are not fully reconstructed; and (iii) the inter-tumor heterogeneity is insufficiently considered. Given that these limitations vary according to the models, preclinical models must be carefully selected depending on the objectives of the experiments. New approaches, such as organ-on-a-chip technologies or in silico biological systems, should be explored in MM research. More pertinent cell models, based on our knowledge on mesothelial carcinogenesis and considering MM heterogeneity, need to be developed. These endeavors are mandatory to implement efficient precision medicine for MM.
Collapse
Affiliation(s)
- Christophe Blanquart
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Marie-Claude Jaurand
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Didier Jean
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors Laboratory, Paris, France
| |
Collapse
|
30
|
Quetel L, Meiller C, Assié JB, Blum Y, Imbeaud S, Montagne F, Tranchant R, de Wolf J, Caruso S, Copin MC, Hofman V, Gibault L, Badoual C, Pintilie E, Hofman P, Monnet I, Scherpereel A, Le Pimpec-Barthes F, Zucman-Rossi J, Jaurand MC, Jean D. Genetic alterations of malignant pleural mesothelioma: association with tumor heterogeneity and overall survival. Mol Oncol 2020; 14:1207-1223. [PMID: 32083805 PMCID: PMC7266286 DOI: 10.1002/1878-0261.12651] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/13/2019] [Accepted: 02/19/2020] [Indexed: 01/17/2023] Open
Abstract
Development of precision medicine for malignant pleural mesothelioma (MPM) requires a deep knowledge of tumor heterogeneity. Histologic and molecular classifications and histo‐molecular gradients have been proposed to describe heterogeneity, but a deeper understanding of gene mutations in the context of MPM heterogeneity is required and the associations between mutations and clinical data need to be refined. We characterized genetic alterations on one of the largest MPM series (266 tumor samples), well annotated with histologic, molecular and clinical data of patients. Targeted next‐generation sequencing was performed focusing on the major MPM mutated genes and the TERT promoter. Molecular heterogeneity was characterized using predictors allowing classification of each tumor into the previously described molecular subtypes and the determination of the proportion of epithelioid‐like and sarcomatoid‐like components (E/S.scores). The mutation frequencies are consistent with literature data, but this study emphasized that TERT promoter, not considered by previous large sequencing studies, was the third locus most affected by mutations in MPM. Mutations in TERT promoter, NF2, and LATS2 were more frequent in nonepithelioid MPM and positively associated with the S.score. BAP1, NF2, TERT promoter, TP53, and SETD2 mutations were enriched in some molecular subtypes. NF2 mutation rate was higher in asbestos unexposed patient. TERT promoter, NF2, and TP53 mutations were associated with a poorer overall survival. Our findings lead to a better characterization of MPM heterogeneity by identifying new significant associations between mutational status and histologic and molecular heterogeneity. Strikingly, we highlight the strong association between new mutations and overall survival.
Collapse
Affiliation(s)
- Lisa Quetel
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Clément Meiller
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Jean-Baptiste Assié
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Yuna Blum
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, Paris, France
| | - Sandrine Imbeaud
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - François Montagne
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Robin Tranchant
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Julien de Wolf
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Marie-Christine Copin
- Institut de Pathologie, Centre de Biologie-Pathologie, CHRU de Lille, France.,Université de Lille, France
| | - Véronique Hofman
- Laboratoire de Pathologie Clinique et Expérimentale (LPCE) et Biobanque (BB-0033-00025), CHRU de Nice, France.,FHU OncoAge, Université Côte d'Azur, Nice, France
| | - Laure Gibault
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France.,Service d'Anatomopathologie et Cytologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Cécile Badoual
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France.,Service d'Anatomopathologie et Cytologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Ecaterina Pintilie
- Service de Chirurgie Thoracique, Hôpital Calmette - CHRU de Lille, France
| | - Paul Hofman
- Laboratoire de Pathologie Clinique et Expérimentale (LPCE) et Biobanque (BB-0033-00025), CHRU de Nice, France.,FHU OncoAge, Université Côte d'Azur, Nice, France
| | - Isabelle Monnet
- Service de Pneumologie et Pathologie Professionnelle, Centre Hospitalier Intercommunal de Créteil, France
| | - Arnaud Scherpereel
- Université de Lille, France.,Service de Pneumologie et d'Oncologie Thoracique, Hôpital Calmette - CHRU de Lille, France.,Réseau National Expert pour le Mésothéliome Pleural Malin (MESOCLIN), Lille, France
| | - Françoise Le Pimpec-Barthes
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France.,Service de Chirurgie Thoracique, Hôpital Européen Georges Pompidou, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Marie-Claude Jaurand
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| | - Didier Jean
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors laboratory, France
| |
Collapse
|
31
|
Testa JR, Berns A. Preclinical Models of Malignant Mesothelioma. Front Oncol 2020; 10:101. [PMID: 32117751 PMCID: PMC7026500 DOI: 10.3389/fonc.2020.00101] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/20/2020] [Indexed: 11/13/2022] Open
Abstract
Rodent models of malignant mesothelioma help facilitate the understanding of the biology of this highly lethal cancer and to develop and test new interventions. Introducing the same genetic lesions as found in human mesothelioma in mice results in tumors that show close resemblance with the human disease counterpart. This includes the extensive inflammatory responses that characterize human malignant mesothelioma. The relatively fast development of mesothelioma in mice when the appropriate combination of lesions is introduced, with or without exposure to asbestos, make the autochthonous models particularly useful for testing new treatment strategies in an immunocompetent setting, whereas Patient-Derived Xenograft models are particularly useful to assess effects of inter- and intra-tumor heterogeneity and human-specific features of mesothelioma. It is to be expected that new insights obtained by studying these experimental systems will lead to new more effective treatments for this devastating disease.
Collapse
Affiliation(s)
- Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
32
|
Abstract
Mesothelioma has long been associated with the exposure to asbestos, which was largely used in manufacturing activities. Toxicology studies in vitro and in vivo demonstrated that asbestos fibers were carcinogenic, and epidemiology studies revealed that asbestos exposure was paralleled by the increase in the incidence of mesothelioma and related mortality rates. More recently, the role of chronic inflammation and the molecular mechanisms involved in carcinogenesis by mineral fibers were elucidated following the discovery of the roles of HMGB1 and inflammasome. A change of paradigm was the discovery of a prevalence of mesotheliomas attributable to inherited mutations of cancer susceptibility genes. The discovery of BAP1 as a predisposition gene for the development of familial mesothelioma and other cancers implemented genome studies in patients with mesothelioma and routine clinical surveys in individuals at risk to identify germline mutations associated with cancers included in the BAP1 syndrome. A further progress in the approach to asbestos-related malignancy was the adoption of combined genetics and environmental analyses according to the model of gene-environment (GxE) interactions. This review aims at updating on the most recently discovered mechanisms of tumorigenesis and the pivotal role of GxE interactions.
Collapse
Affiliation(s)
| | - Jiaming Xue
- University of Hawai'i Cancer Center, Honolulu, HI 96813, USA
| | - Haining Yang
- University of Hawai'i Cancer Center, Honolulu, HI 96813, USA
| |
Collapse
|
33
|
Malouf GG, Flippot R, Dong Y, Dinatale RG, Chen YB, Su X, Compérat E, Rouprêt M, Mano R, Blum KA, Yao H, Mouawad R, Spano JP, Khayat D, Karam JA, Ho TH, Tickoo SK, Russo P, Hsieh JJ, Tannir NM, Hakimi AA. Molecular characterization of sarcomatoid clear cell renal cell carcinoma unveils new candidate oncogenic drivers. Sci Rep 2020; 10:701. [PMID: 31959902 PMCID: PMC6971072 DOI: 10.1038/s41598-020-57534-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/17/2019] [Indexed: 11/18/2022] Open
Abstract
Sarcomatoid clear-cell renal cell carcinomas (sRCC) are associated with dismal prognosis. Genomic alterations associated with sarcomatoid dedifferentiation are poorly characterized. We sought to define the genomic landscape of sRCC and uncover potentially actionable therapeutic targets. We assessed the genomic landscape of sRCC using targeted panel sequencing including patients with microdissected sarcomatoid and epithelial components. Along with common genomic alterations associated with clear-cell histology, we found that Hippo was one of the most frequently altered pathways in these tumours. Hippo alterations were differentially enriched in sRCC compared to non-sRCC. Functional analysis showed that Hippo members mutations were associated with higher nuclear accumulation of YAP/TAZ, core effectors of the Hippo pathway. In a NF2-mutant sRCC model, YAP1 knockdown and NF2 reconstitution suppressed cell proliferation, tumour growth and invasion, both in vitro and in vivo. Overall, we show that Hippo pathway alterations are a feature of sRCC, and enable the exploration of the Hippo pathway as a novel potential therapeutic target.
Collapse
Affiliation(s)
- Gabriel G Malouf
- Department of Medical Oncology, Hôpital Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France. .,Department of Hematology and Oncology, Centre Hospitalier Universitaire Régional de Strasbourg, Institut de Cancérologie de Strasbourg, Strasbourg, France. .,Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, Illkirch, France.
| | - Ronan Flippot
- Department of Medical Oncology, Hôpital Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Yiyu Dong
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renzo G Dinatale
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaoping Su
- Department of Biostatistics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eva Compérat
- Department of Pathology, Hôpital Tenon, APHP, Sorbonne Université, Paris, France
| | - Morgan Rouprêt
- Department of Urology, Hôpital Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Roy Mano
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyle A Blum
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hui Yao
- Department of Biostatistics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roger Mouawad
- Department of Medical Oncology, Hôpital Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Jean-Philippe Spano
- Department of Medical Oncology, Hôpital Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France.,Inserm UMRS 1136, Sorbonne Université, Paris, France
| | - David Khayat
- Department of Medical Oncology, Hôpital Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Jose A Karam
- Department of Urology, MD Anderson Cancer Center, Houston, TX, USA
| | - Thai H Ho
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Epigenomics Group, Mayo Clinic, Rochester, MN, USA
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Russo
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James J Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO, USA
| | - Nizar M Tannir
- Department of Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Abraham A Hakimi
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
34
|
Marqués M, Tranchant R, Risa-Ebrí B, Suárez-Solís ML, Fernández LC, Carrillo-de-Santa-Pau E, Del Pozo N, Martínez de Villarreal J, Meiller C, Allory Y, Blum Y, Pirker C, Hegedus B, Barry ST, Carnero A, Berger W, Jean D, Real FX. Combined MEK and PI3K/p110β Inhibition as a Novel Targeted Therapy for Malignant Mesothelioma Displaying Sarcomatoid Features. Cancer Res 2020; 80:843-856. [PMID: 31911549 DOI: 10.1158/0008-5472.can-19-1633] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/01/2019] [Accepted: 12/18/2019] [Indexed: 11/16/2022]
Abstract
Among malignant mesotheliomas (MM), the sarcomatoid subtype is associated with higher chemoresistance and worst survival. Due to its low incidence, there has been little progress in the knowledge of the molecular mechanisms associated with sarcomatoid MM, which might help to define novel therapeutic targets. In this work, we show that loss of PTEN expression is frequent in human sarcomatoid MM and PTEN expression levels are lower in sarcomatoid MM than in the biphasic and epithelioid subtypes. Combined Pten and Trp53 deletion in mouse mesothelium led to nonepithelioid MM development. In Pten;Trp53-null mice developing MM, the Gαi2-coupled receptor subunit activated MEK/ERK and PI3K, resulting in aggressive, immune-suppressed tumors. Combined inhibition of MEK and p110β/PI3K reduced mouse tumor cell growth in vitro. Therapeutic inhibition of MEK and p110β/PI3K using selumetinib (AZD6244, ARRY-142886) and AZD8186, two drugs that are currently in clinical trials, increased the survival of Pten;Trp53-null mice without major toxicity. This drug combination effectively reduced the proliferation of primary cultures of human pleural (Pl) MM, implicating nonepithelioid histology and high vimentin, AKT1/2, and Gαi2 expression levels as predictive markers of response to combined MEK and p110β/PI3K inhibition. Our findings provide a rationale for the use of selumetinib and AZD8186 in patients with MM with sarcomatoid features. This constitutes a novel targeted therapy for a poor prognosis and frequently chemoresistant group of patients with MM, for whom therapeutic options are currently lacking. SIGNIFICANCE: Mesothelioma is highly aggressive; its sarcomatoid variants have worse prognosis. Building on a genetic mouse model, a novel combination therapy is uncovered that is relevant to human tumors.
Collapse
Affiliation(s)
- Miriam Marqués
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain. .,CIBERONC, Madrid, Spain
| | - Robin Tranchant
- Centre de Recherche des Cordeliers, INSERM, Université Paris Descartes, Université Paris Diderot, Sorbonne Université, USPC, Functional Genomics of Solid Tumors Team, Paris, France
| | - Blanca Risa-Ebrí
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain
| | - María L Suárez-Solís
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain.,Department of Surgical Pathology, Hospital Clínico San Carlos, Madrid, Spain
| | - Luis C Fernández
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain.,Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, Madrid, Spain
| | - Enrique Carrillo-de-Santa-Pau
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain.,Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, Madrid, Spain
| | - Natalia Del Pozo
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain.,CIBERONC, Madrid, Spain
| | | | - Clément Meiller
- Centre de Recherche des Cordeliers, INSERM, Université Paris Descartes, Université Paris Diderot, Sorbonne Université, USPC, Functional Genomics of Solid Tumors Team, Paris, France
| | - Yves Allory
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain.,Université Paris-Est Créteil, France INSERM, U955, Institut Mondor de Recherche Biomédicales AP-HP, Hôpital Henri Mondor, Department of Pathology, Créteil, France
| | - Yuna Blum
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, Paris, France
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Balazs Hegedus
- Department of Thoracic Surgery, Medical of University Vienna, Vienna, Austria
| | - Simon T Barry
- IMED Oncology, AstraZeneca, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Amancio Carnero
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain.,CIBERONC, Madrid, Spain.,Instituto de Biomedicina de Sevilla, IBIS/HUVR/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Didier Jean
- Centre de Recherche des Cordeliers, INSERM, Université Paris Descartes, Université Paris Diderot, Sorbonne Université, USPC, Functional Genomics of Solid Tumors Team, Paris, France
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain. .,CIBERONC, Madrid, Spain.,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
35
|
Hinz TK, Heasley LE. Translating mesothelioma molecular genomics and dependencies into precision oncology-based therapies. Semin Cancer Biol 2019; 61:11-22. [PMID: 31546009 DOI: 10.1016/j.semcancer.2019.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/28/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare, yet lethal asbestos-induced cancer and despite marked efforts to reduce occupational exposure, the incidence has not yet significantly declined. Since 2003, combined treatment with a platinum-based agent and pemetrexed has been the first-line therapy and no effective or approved second-line treatments have emerged. The seemingly slow advance in developing new MPM treatments does not appear to be related to a low level of clinical and pre-clinical research activity. Rather, we suggest that a key hurdle in successfully translating basic discovery into novel MPM therapeutics is the underlying assumption that as a rare cancer, it will also be molecularly and genetically homogeneous. In fact, lung adenocarcinoma and melanoma only benefitted from precision oncology upon full appreciation of the high degree of molecular heterogeneity inherent in these cancers, especially regarding the diversity of oncogenic drivers. Herein, we consider the recent explosion of molecular and genetic information that has become available regarding MPM and suggest ways in which the unfolding landscape may guide identification of novel therapeutic vulnerabilities within subsets of MPM that can be targeted in a manner consistent with the tenets of precision oncology.
Collapse
Affiliation(s)
- Trista K Hinz
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Lynn E Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
36
|
Kukuyan AM, Sementino E, Kadariya Y, Menges CW, Cheung M, Tan Y, Cai KQ, Slifker MJ, Peri S, Klein-Szanto AJ, Rauscher FJ, Testa JR. Inactivation of Bap1 Cooperates with Losses of Nf2 and Cdkn2a to Drive the Development of Pleural Malignant Mesothelioma in Conditional Mouse Models. Cancer Res 2019; 79:4113-4123. [PMID: 31151962 DOI: 10.1158/0008-5472.can-18-4093] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/01/2019] [Accepted: 05/30/2019] [Indexed: 12/21/2022]
Abstract
Pleural malignant mesothelioma is a therapy-resistant cancer affecting the serosal lining of the thoracic cavity. Mutations/deletions of BAP1, CDKN2A, and NF2 are the most frequent genetic lesions in human malignant mesothelioma. We introduced various combinations of these deletions in the pleura of conditional knockout (CKO) mice, focusing on the contribution of Bap1 loss. While homozygous CKO of Bap1, Cdkn2a, or Nf2 alone gave rise to few or no malignant mesotheliomas, inactivation of Bap1 cooperated with loss of either Nf2 or Cdkn2a to drive development of malignant mesothelioma in approximately 20% of double-CKO mice, and a high incidence (22/26, 85%) of malignant mesotheliomas was observed in Bap1;Nf2;Cdkn2a (triple)-CKO mice. Malignant mesothelioma onset was rapid in triple-CKO mice, with a median survival of only 12 weeks, and malignant mesotheliomas from these mice were consistently high-grade and invasive. Adenoviral-Cre treatment of normal mesothelial cells from Bap1;Nf2;Cdkn2a CKO mice, but not from mice with knockout of one or any two of these genes, resulted in robust spheroid formation in vitro, suggesting that mesothelial cells from Bap1;Nf2;Cdkn2a mice have stem cell-like potential. RNA-seq analysis of malignant mesotheliomas from triple-CKO mice revealed enrichment of genes transcriptionally regulated by the polycomb repressive complex 2 (PRC2) and others previously implicated in known Bap1-related cellular processes. These data demonstrate that somatic inactivation of Bap1, Nf2, and Cdkn2a results in rapid, aggressive malignant mesotheliomas, and that deletion of Bap1 contributes to tumor development, in part, by loss of PRC2-mediated repression of tumorigenic target genes and by acquisition of stem cell potential, suggesting a potential avenue for therapeutic intervention. SIGNIFICANCE: Combinatorial deletions of Bap1, Nf2, and Cdkn2a result in aggressive mesotheliomas, with Bap1 loss contributing to tumorigenesis by circumventing PRC2-mediated repression of oncogenic target genes.
Collapse
Affiliation(s)
| | - Eleonora Sementino
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yuwaraj Kadariya
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Craig W Menges
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mitchell Cheung
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yinfei Tan
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Michael J Slifker
- Bioinformatics and Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Suraj Peri
- Bioinformatics and Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | | | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
37
|
Tian K, Bakker E, Hussain M, Guazzelli A, Alhebshi H, Meysami P, Demonacos C, Schwartz JM, Mutti L, Krstic-Demonacos M. p53 modeling as a route to mesothelioma patients stratification and novel therapeutic identification. J Transl Med 2018; 16:282. [PMID: 30316293 PMCID: PMC6186085 DOI: 10.1186/s12967-018-1650-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/01/2018] [Indexed: 12/16/2022] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is an orphan disease that is difficult to treat using traditional chemotherapy, an approach which has been effective in other types of cancer. Most chemotherapeutics cause DNA damage leading to cell death. Recent discoveries have highlighted a potential role for the p53 tumor suppressor in this disease. Given the pivotal role of p53 in the DNA damage response, here we investigated the predictive power of the p53 interactome model for MPM patients’ stratification. Methods We used bioinformatics approaches including omics type analysis of data from MPM cells and from MPM patients in order to predict which pathways are crucial for patients’ survival. Analysis of the PKT206 model of the p53 network was validated by microarrays from the Mero-14 MPM cell line and RNA-seq data from 71 MPM patients, whilst statistical analysis was used to identify the deregulated pathways and predict therapeutic schemes by linking the affected pathway with the patients’ clinical state. Results In silico simulations demonstrated successful predictions ranging from 52 to 85% depending on the drug, algorithm or sample used for validation. Clinical outcomes of individual patients stratified in three groups and simulation comparisons identified 30 genes that correlated with survival. In patients carrying wild-type p53 either treated or not treated with chemotherapy, FEN1 and MMP2 exhibited the highest inverse correlation, whereas in untreated patients bearing mutated p53, SIAH1 negatively correlated with survival. Numerous repositioned and experimental drugs targeting FEN1 and MMP2 were identified and selected drugs tested. Epinephrine and myricetin, which target FEN1, have shown cytotoxic effect on Mero-14 cells whereas marimastat and batimastat, which target MMP2 demonstrated a modest but significant inhibitory effect on MPM cell migration. Finally, 8 genes displayed correlation with disease stage, which may have diagnostic implications. Conclusions Clinical decisions related to MPM personalized therapy based on individual patients’ genetic profile and previous chemotherapeutic treatment could be reached using computational tools and the predictions reported in this study upon further testing in animal models. Electronic supplementary material The online version of this article (10.1186/s12967-018-1650-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kun Tian
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Emyr Bakker
- School of Medicine, University of Central Lancashire, Preston, UK
| | | | - Alice Guazzelli
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Hasen Alhebshi
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Parisa Meysami
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | | | - Jean-Marc Schwartz
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | | |
Collapse
|
38
|
Felley-Bosco E, MacFarlane M. Asbestos: Modern Insights for Toxicology in the Era of Engineered Nanomaterials. Chem Res Toxicol 2018; 31:994-1008. [PMID: 30156102 DOI: 10.1021/acs.chemrestox.8b00146] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Asbestos fibers are naturally occurring silicates that have been extensively used in the past, including house construction, but because of their toxicity, their use has been banned in 63 countries. Despite this, more than one million metric tons of asbestos are still consumed annually in countries where asbestos use has not been banned. Asbestos-related disease incidence is still increasing in several countries, including those countries that banned the use of asbestos more than 30 years ago. We highlight here recent knowledge obtained in experimental models about the mechanisms leading to tumor development following asbestos exposure, including genetic and epigenetic changes. Importantly, the landscape of alterations observed experimentally in tumor samples is consistent with alterations observed in clinical tumor samples; therefore, studies performed on early/precancer stages should help inform secondary prevention, which remains crucial in the absence of an efficient primary prevention. Knowledge gathered on asbestos should also help address future challenges, especially in view of the increased production of new materials that may behave similarly to asbestos fibers.
Collapse
Affiliation(s)
- Emanuela Felley-Bosco
- Laboratory of Molecular Oncology , University Hospital Zurich , Sternwartstrasse 14 , 8091 Zürich , Switzerland
| | - Marion MacFarlane
- MRC Toxicology Unit , University of Cambridge , Hodgkin Building, Leicester LE1 9HN , United Kingdom
| |
Collapse
|
39
|
Jean D, Jaurand MC. Mesotheliomas in Genetically Engineered Mice Unravel Mechanism of Mesothelial Carcinogenesis. Int J Mol Sci 2018; 19:E2191. [PMID: 30060470 PMCID: PMC6121615 DOI: 10.3390/ijms19082191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
Malignant mesothelioma (MM), a rare and severe cancer, mainly caused as a result of past-asbestos exposure, is presently a public health concern. Current molecular studies aim to improve the outcome of the disease, providing efficient therapies based on the principles of precision medicine. To model the molecular profile of human malignant mesothelioma, animal models have been developed in rodents, wild type animals and genetically engineered mice harbouring mutations in tumour suppressor genes, especially selecting genes known to be inactivated in human malignant mesothelioma. Animals were either exposed or not exposed to asbestos or to other carcinogenic fibres, to understand the mechanism of action of fibres at the molecular level, and the role of the selected genes in mesothelial carcinogenesis. The aim of the manuscript was to compare mesothelioma models to human malignant mesothelioma and to specify the clue genes playing a role in mesothelial carcinogenesis. Collectively, MM models recapitulate the clinical features of human MM. At least two altered genes are needed to induce malignant mesothelioma in mice. Two pathways regulated by Cdkn2a and Trp53 seem independent key players in mesothelial carcinogenesis. Other genes and pathways appear as bona fide modulators of the neoplastic transformation.
Collapse
Affiliation(s)
- Didier Jean
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, F-75010 Paris, France.
- Université Paris Descartes, Labex Immuno-Oncologie, Sorbonne Paris Cité, F-75000 Paris, France.
- Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, F-75010 Paris, France.
- Université Paris 13, Sorbonne Paris Cité, F-93206 Saint-Denis, France.
| | - Marie-Claude Jaurand
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, F-75010 Paris, France.
- Université Paris Descartes, Labex Immuno-Oncologie, Sorbonne Paris Cité, F-75000 Paris, France.
- Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, F-75010 Paris, France.
- Université Paris 13, Sorbonne Paris Cité, F-93206 Saint-Denis, France.
| |
Collapse
|
40
|
Nabavi N, Wei J, Lin D, Collins CC, Gout PW, Wang Y. Pre-clinical Models for Malignant Mesothelioma Research: From Chemical-Induced to Patient-Derived Cancer Xenografts. Front Genet 2018; 9:232. [PMID: 30022998 PMCID: PMC6040159 DOI: 10.3389/fgene.2018.00232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/11/2018] [Indexed: 01/19/2023] Open
Abstract
Malignant mesothelioma (MM) is a rare disease often associated with environmental exposure to asbestos and other erionite fibers. MM has a long latency period prior to manifestation and a poor prognosis. The survival post-diagnosis is often less than a year. Although use of asbestos has been banned in the United States and many European countries, asbestos is still being used and extracted in many developing countries. Occupational exposure to asbestos, mining, and migration are reasons that we expect to continue to see growing incidence of mesothelioma in the coming decades. Despite improvements in survival achieved with multimodal therapies and cytoreductive surgeries, less morbid, more effective interventions are needed. Thus, identifying prognostic and predictive biomarkers for MM, and developing novel agents for targeted therapy, are key unmet needs in mesothelioma research and treatment. In this review, we discuss the evolution of pre-clinical model systems developed to study MM and emphasize the remarkable capability of patient-derived xenograft (PDX) MM models in expediting the pre-clinical development of novel therapeutic approaches. PDX disease model systems retain major characteristics of original malignancies with high fidelity, including molecular, histopathological and functional heterogeneities, and as such play major roles in translational research, drug development, and precision medicine.
Collapse
Affiliation(s)
- Noushin Nabavi
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Jingchao Wei
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Department of Urology, the Third Xiangya Hospital, Central South University Changsha, China
| | - Dong Lin
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Colin C Collins
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Peter W Gout
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| |
Collapse
|
41
|
Sementino E, Menges CW, Kadariya Y, Peri S, Xu J, Liu Z, Wilkes RG, Cai KQ, Rauscher FJ, Klein-Szanto AJ, Testa JR. Inactivation of Tp53 and Pten drives rapid development of pleural and peritoneal malignant mesotheliomas. J Cell Physiol 2018; 233:8952-8961. [PMID: 29904909 DOI: 10.1002/jcp.26830] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/09/2018] [Indexed: 02/03/2023]
Abstract
Malignant mesothelioma (MM) is a therapy-resistant cancer arising primarily from the lining of the pleural and peritoneal cavities. The most frequently altered genes in human MM are cyclin-dependent kinase inhibitor 2A (CDKN2A), which encodes components of the p53 (p14ARF) and RB (p16INK4A) pathways, BRCA1-associated protein 1 (BAP1), and neurofibromatosis 2 (NF2). Furthermore, the p53 gene (TP53) itself is mutated in ~15% of MMs. In many MMs, the PI3K-PTEN-AKT-mTOR signaling node is hyperactivated, which contributes to tumor cell survival and therapeutic resistance. Here, we demonstrate that the inactivation of both Tp53 and Pten in the mouse mesothelium is sufficient to rapidly drive aggressive MMs. PtenL/L ;Tp53L/L mice injected intraperitoneally or intrapleurally with adenovirus-expressing Cre recombinase developed high rates of peritoneal and pleural MMs (92% of mice with a median latency of 9.4 weeks and 56% of mice with a median latency of 19.3 weeks, respectively). MM cells from these mice showed consistent activation of Akt-mTor signaling, chromosome breakage or aneuploidy, and upregulation of Myc; occasional downregulation of Bap1 was also observed. Collectively, these findings suggest that when Pten and Tp53 are lost in combination in mesothelial cells, DNA damage is not adequately repaired and genomic instability is widespread, whereas the activation of Akt due to Pten loss protects genomically damaged cells from apoptosis, thereby increasing the likelihood of tumor formation. Additionally, the mining of an online dataset (The Cancer Genome Atlas) revealed codeletions of PTEN and TP53 and/or CDKN2A/p14ARF in ~25% of human MMs, indicating that cooperative losses of these genes contribute to the development of a significant proportion of these aggressive neoplasms and suggesting key target pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Eleonora Sementino
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Craig W Menges
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yuwaraj Kadariya
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Suraj Peri
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Jinfei Xu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Zemin Liu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Richard G Wilkes
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Frank J Rauscher
- Gene Expression and Regulation Program, Wistar Institute, Philadelphia, Pennsylvania
| | | | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
42
|
Felley-Bosco E, Rehrauer H. Non-Coding Transcript Heterogeneity in Mesothelioma: Insights from Asbestos-Exposed Mice. Int J Mol Sci 2018; 19:ijms19041163. [PMID: 29641489 PMCID: PMC5979355 DOI: 10.3390/ijms19041163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/05/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022] Open
Abstract
Mesothelioma is an aggressive, rapidly fatal cancer and a better understanding of its molecular heterogeneity may help with making more efficient therapeutic strategies. Non-coding RNAs represent a larger part of the transcriptome but their contribution to diseases is not fully understood yet. We used recently obtained RNA-seq data from asbestos-exposed mice and performed data mining of publicly available datasets in order to evaluate how non-coding RNA contribute to mesothelioma heterogeneity. Nine non-coding RNAs are specifically elevated in mesothelioma tumors and contribute to human mesothelioma heterogeneity. Because some of them have known oncogenic properties, this study supports the concept of non-coding RNAs as cancer progenitor genes.
Collapse
Affiliation(s)
- Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Lungen- und Thoraxonkologie Zentrum, University Hospital Zurich, Sternwartstrasse 14, 8091 Zürich, Switzerland.
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
43
|
NF2/Merlin Inactivation and Potential Therapeutic Targets in Mesothelioma. Int J Mol Sci 2018; 19:ijms19040988. [PMID: 29587439 PMCID: PMC5979333 DOI: 10.3390/ijms19040988] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
The neurofibromatosis type 2 (NF2) gene encodes merlin, a tumor suppressor protein frequently inactivated in schwannoma, meningioma, and malignant mesothelioma (MM). The sequence of merlin is similar to that of ezrin/radixin/moesin (ERM) proteins which crosslink actin with the plasma membrane, suggesting that merlin plays a role in transducing extracellular signals to the actin cytoskeleton. Merlin adopts a distinct closed conformation defined by specific intramolecular interactions and regulates diverse cellular events such as transcription, translation, ubiquitination, and miRNA biosynthesis, many of which are mediated through Hippo and mTOR signaling, which are known to be closely involved in cancer development. MM is a very aggressive tumor associated with asbestos exposure, and genetic alterations in NF2 that abrogate merlin’s functional activity are found in about 40% of MMs, indicating the importance of NF2 inactivation in MM development and progression. In this review, we summarize the current knowledge of molecular events triggered by NF2/merlin inactivation, which lead to the development of mesothelioma and other cancers, and discuss potential therapeutic targets in merlin-deficient mesotheliomas.
Collapse
|
44
|
Targeting the Hippo Pathway Is a New Potential Therapeutic Modality for Malignant Mesothelioma. Cancers (Basel) 2018; 10:cancers10040090. [PMID: 29565815 PMCID: PMC5923345 DOI: 10.3390/cancers10040090] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 12/14/2022] Open
Abstract
Malignant mesothelioma (MM) constitutes a very aggressive tumor that arises from the pleural or peritoneal cavities and is highly refractory to conventional therapies. Several key genetic alterations are associated with the development and progression of MM including mutations of the CDKN2A/ARF, NF2, and BAP1 tumor-suppressor genes. Notably, activating oncogene mutations are very rare; thus, it is difficult to develop effective inhibitors to treat MM. The NF2 gene encodes merlin, a protein that regulates multiple cell-signaling cascades including the Hippo pathway. MMs also exhibit inactivation of Hippo pathway components including LATS1/2, strongly suggesting that merlin-Hippo pathway dysregulation plays a key role in the development and progression of MM. Furthermore, Hippo pathway inactivation has been shown to result in constitutive activation of the YAP1/TAZ transcriptional coactivators, thereby conferring malignant phenotypes to mesothelial cells. Critical YAP1/TAZ target genes, including prooncogenic CCDN1 and CTGF, have also been shown to enhance the malignant phenotypes of MM cells. Together, these data indicate the Hippo pathway as a therapeutic target for the treatment of MM, and support the development of new strategies to effectively target the activation status of YAP1/TAZ as a promising therapeutic modality for this formidable disease.
Collapse
|
45
|
Rehrauer H, Wu L, Blum W, Pecze L, Henzi T, Serre-Beinier V, Aquino C, Vrugt B, de Perrot M, Schwaller B, Felley-Bosco E. How asbestos drives the tissue towards tumors: YAP activation, macrophage and mesothelial precursor recruitment, RNA editing, and somatic mutations. Oncogene 2018; 37:2645-2659. [PMID: 29507420 PMCID: PMC5955862 DOI: 10.1038/s41388-018-0153-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/11/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022]
Abstract
Chronic exposure to intraperitoneal asbestos triggered a marked response in the mesothelium well before tumor development. Macrophages, mesothelial precursor cells, cytokines, and growth factors accumulated in the peritoneal lavage. Transcriptome profiling revealed YAP/TAZ activation in inflamed mesothelium with further activation in tumors, paralleled by increased levels of cells with nuclear YAP/TAZ. Arg1 was one of the highest upregulated genes in inflamed tissue and tumor. Inflamed tissue showed increased levels of single-nucleotide variations, with an RNA-editing signature, which were even higher in the tumor samples. Subcutaneous injection of asbestos-treated, but tumor-free mice with syngeneic mesothelioma tumor cells resulted in a significantly higher incidence of tumor growth when compared to naïve mice supporting the role of the environment in tumor progression.
Collapse
Affiliation(s)
- Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital University Health Network, University of Toronto, Toronto, ON, Canada
| | - Walter Blum
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Lazslo Pecze
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Thomas Henzi
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | | | - Catherine Aquino
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Bart Vrugt
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital University Health Network, University of Toronto, Toronto, ON, Canada
| | - Beat Schwaller
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Lungen- und Thoraxonkologie Zentrum, University Hospital Zürich, Sternwartstrasse 14, 8091, Zurich, Switzerland.
| |
Collapse
|
46
|
Oehl K, Kresoja-Rakic J, Opitz I, Vrugt B, Weder W, Stahel R, Wild P, Felley-Bosco E. Live-Cell Mesothelioma Biobank to Explore Mechanisms of Tumor Progression. Front Oncol 2018. [PMID: 29527515 PMCID: PMC5829086 DOI: 10.3389/fonc.2018.00040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Experimental models closely representing in vivo conditions allow investigating mechanisms of resistance. Our aims were to establish a live-cell biobank of malignant pleural mesothelioma (MPM) samples and to obtain proof of principle that primary culture chemoresistant models, mimicking tumor progression observed in patients, can be obtained in vitro, providing a useful tool to investigate underlying mechanisms. Primary mesothelioma cultures were established from 235 samples between 2007 and 2014. Of two MPM patients, primary cultures obtained at different time points: at initial diagnosis, after neoadjuvant treatment at surgery and/or after tumor recurrence, were deeply investigated. Cells and corresponding tumor tissue were characterized by mesothelial protein and gene expression analysis. In addition, primary cultures from chemo naive patients were exposed to increasing doses of cisplatin/pemetrexed during three months and compared with non-treated cells in a cytotoxicity assay, and by selected profiling of senescence markers. In vitro chemoresistance in the primary mesothelioma cell cultures was associated with increased Thy1 (CD90) expression. Thy1 expression in MPM samples was significantly associated with poor overall survival in the TCGA MPM cohort. Our results illustrate that the establishment of a large live-cell MPM biobank contributes to a better understanding of therapy resistance observed in vivo, which eventually may lead to a more logical approach for developing new treatment strategies.
Collapse
Affiliation(s)
- Kathrin Oehl
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Jelena Kresoja-Rakic
- Laboratory of Molecular Oncology, Division of Thoracic Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Isabelle Opitz
- Laboratory of Molecular Oncology, Division of Thoracic Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Bart Vrugt
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Walter Weder
- Laboratory of Molecular Oncology, Division of Thoracic Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Rolf Stahel
- Cancer Center Zürich, University Hospital Zürich, Zürich, Switzerland
| | - Peter Wild
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Division of Thoracic Surgery, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
47
|
Parrotta R, Okonska A, Ronner M, Weder W, Stahel R, Penengo L, Felley-Bosco E. A Novel BRCA1-Associated Protein-1 Isoform Affects Response of Mesothelioma Cells to Drugs Impairing BRCA1-Mediated DNA Repair. J Thorac Oncol 2017; 12:1309-1319. [DOI: 10.1016/j.jtho.2017.03.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 03/08/2017] [Accepted: 03/21/2017] [Indexed: 01/05/2023]
|
48
|
Abstract
Malignant mesothelioma is a universally lethal cancer that is increasing in incidence worldwide. There is a dearth of effective therapies, with only one treatment (pemetrexed and cisplatin combination chemotherapy) approved in the past 13 years. However, the past 5 years have witnessed an exponential growth in our understanding of mesothelioma pathobiology, which is set to revolutionize therapeutic strategies. From a genomic standpoint, mesothelioma is characterized by a preponderance of tumour suppressor alterations, for which novel therapies are currently in development. Other promising antitumour agents include inhibitors against angiogenesis, mesothelin and immune checkpoints, which are at various phases of clinical trial testing.
Collapse
Affiliation(s)
- Timothy A Yap
- The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Joachim G Aerts
- Erasmus MC Cancer Institute, 3015 CE Rotterdam, The Netherlands
| | - Sanjay Popat
- Royal Marsden Hospital, London SW3 6JJ, UK
- National Heart and Lung Institute, Imperial College London SW3 6NP, UK
| | | |
Collapse
|
49
|
Psallidas I, Kalomenidis I, Porcel JM, Robinson BW, Stathopoulos GT. Malignant pleural effusion: from bench to bedside. Eur Respir Rev 2017; 25:189-98. [PMID: 27246596 DOI: 10.1183/16000617.0019-2016] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/25/2016] [Indexed: 11/05/2022] Open
Abstract
Malignant pleural effusion (MPE) is a common but serious condition that is related with poor quality of life, morbidity and mortality. Its incidence and associated healthcare costs are rising and its management remains palliative, with median survival ranging from 3 to 12 months. During the last decade there has been significant progress in unravelling the pathophysiology of MPE, as well as its diagnostics, imaging, and management. Nowadays, formerly bed-ridden patients are genotyped, phenotyped, and treated on an ambulatory basis. This article attempts to provide a comprehensive overview of current advances in MPE from bench to bedside. In addition, it highlights unanswered questions in current clinical practice and suggests future directions for basic and clinical research in the field.
Collapse
Affiliation(s)
- Ioannis Psallidas
- Oxford Respiratory Trials Unit, Oxford Centre for Respiratory Medicine, Oxford University Hospitals Trust, Oxford, UK
| | - Ioannis Kalomenidis
- 1st Dept of Critical Care and Pulmonary Medicine, National and Kapodistrian University of Athens, School of Medicine, Evangelismos Hospital, Athens, Greece
| | - Jose M Porcel
- Pleural Medicine Unit, Dept of Internal Medicine, Arnau de Vilanova University Hospital, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Bruce W Robinson
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia Dept of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Achaia, Greece Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
50
|
Tranchant R, Quetel L, Tallet A, Meiller C, Renier A, de Koning L, de Reynies A, Le Pimpec-Barthes F, Zucman-Rossi J, Jaurand MC, Jean D. Co-occurring Mutations of Tumor Suppressor Genes, LATS2 and NF2, in Malignant Pleural Mesothelioma. Clin Cancer Res 2016; 23:3191-3202. [PMID: 28003305 DOI: 10.1158/1078-0432.ccr-16-1971] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/18/2016] [Accepted: 12/09/2016] [Indexed: 11/16/2022]
Abstract
Purpose: To better define malignant pleural mesothelioma (MPM) heterogeneity and identify molecular subtypes of MPM, we focus on the tumor suppressor gene LATS2, a member of the Hippo signaling pathway, which plays a key role in mesothelial carcinogenesis.Experimental Design: Sixty-one MPM primary cultures established in our laboratory were screened for mutations in LATS2 Gene inactivation was modeled using siRNAs. Gene and protein expressions were analyzed by quantitative RT-PCR, Western blot analysis, and reverse phase protein array. Cell proliferation, viability, apoptosis, mobility, and invasion were determined after siRNA knockdown or YAP (verteporfin), mTOR (rapamycin), and mTOR/PI3K/AKT (PF-04691502) inhibitor treatment.Results: The LATS2 gene was altered in 11% of MPM by point mutations and large exon deletions. Genetic data coupled with transcriptomic data allowed the identification of a new MPM molecular subgroup, C2LN, characterized by a co-occurring mutation in the LATS2 and NF2 genes in the same MPM. MPM patients of this subgroup presented a poor prognosis. Coinactivation of LATS2 and NF2 leads to loss of cell contact inhibition between MPM cells. Hippo signaling pathway activity, mTOR expression, and phosphorylation were altered in the C2LN MPM subgroup. MPMs of this new subgroup show higher sensitivity to PF-04691502 inhibitor. The MOK gene was identified as a potential biomarker of the C2LN MPM subgroup and PF-04691502 sensitivity.Conclusions: We identified a new MPM molecular subgroup that shares common genetic and transcriptomic characteristics. Our results made it possible to highlight a greater sensitivity to an anticancer compound for this MPM subgroup and to identify a specific potential biomarker. Clin Cancer Res; 23(12); 3191-202. ©2016 AACR.
Collapse
Affiliation(s)
- Robin Tranchant
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France
| | - Lisa Quetel
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France
| | - Anne Tallet
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France
| | - Clement Meiller
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France
| | - Annie Renier
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France
| | - Leanne de Koning
- Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Aurelien de Reynies
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, Paris, France
| | - Francoise Le Pimpec-Barthes
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France.,Département de Chirurgie Thoracique, Hopital Européen Georges Pompidou, Paris, France.,Assistance Publique-Hopitaux de Paris, Hopital Européen Georges Pompidou, Paris, France
| | - Jessica Zucman-Rossi
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France.,Assistance Publique-Hopitaux de Paris, Hopital Européen Georges Pompidou, Paris, France
| | - Marie-Claude Jaurand
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France
| | - Didier Jean
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR-1162, Equipe labellisée Ligue Contre le Cancer, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Labex Immuno-oncology, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Saint-Denis, France
| |
Collapse
|