G. Zalcman, J. Mazieres, J. Margery, L. Greillier, C. Audigier-valette et al., Bevacizumab for newly diagnosed pleural mesothelioma in the mesothelioma avastin cisplatin pemetrexed study (MAPS): a randomised, controlled, open-label, phase 3 trial, Lancet, vol.387, pp.1405-1419, 2016.

L. Calabro, A. Morra, E. Fonsatti, O. Cutaia, G. Amato et al., Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial, Lancet Oncol, vol.14, pp.1104-1115, 2013.

M. Maio, A. Scherpereel, L. Calabro, J. Aerts, C. Perez et al., Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): a multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial, Lancet Oncol, vol.18, issue.17, pp.30446-30447, 2017.

A. Scherpereel, J. Mazieres, L. Greillier, S. Lantuejoul, P. Do et al., Nivolumab or nivolumab plus ipilimumab in patients with relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): a multicentre, openlabel, randomised, non-comparative, phase 2 trial, Lancet Oncol, vol.20, pp.239-53, 2019.

P. S. Adusumilli, L. Cherkassky, J. Villena-vargas, C. Colovos, E. Servais et al., Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity, Sci Transl Med, vol.6, pp.261-151, 2014.

J. Aerts, P. L. De-goeje, R. Cornelissen, M. Kaijen-lambers, K. Bezemer et al., Autologous dendritic cells pulsed with allogeneic tumor cell lysate in mesothelioma: from mouse to human, Clin Cancer Res, vol.24, pp.766-76, 2018.

G. L. Beatty, A. R. Haas, M. V. Maus, D. A. Torigian, M. C. Soulen et al., Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies, Cancer Immunol Res, vol.2, pp.112-132, 2014.

R. A. Belderbos, P. Baas, R. Berardi, R. Cornelissen, D. A. Fennell et al., A multicenter, randomized, phase II/III study of dendritic cells loaded with allogeneic tumor cell lysate (MesoPher) in subjects with mesothelioma as maintenance therapy after chemotherapy: DENdritic cell immunotherapy for mesothelioma (DENIM) trial, Transl Lung Cancer Res, vol.8, pp.280-285, 2019.

R. Cornelissen, J. P. Hegmans, A. P. Maat, M. E. Kaijen-lambers, K. Bezemer et al., Extended tumor control after dendritic cell vaccination with low-dose cyclophosphamide as adjuvant treatment in patients with malignant pleural mesothelioma, Am J Respir Crit Care Med, vol.193, pp.1023-1054, 2016.

J. P. Hegmans, J. D. Veltman, M. E. Lambers, I. J. De-vries, C. G. Figdor et al., Consolidative dendritic cell-based immunotherapy elicits cytotoxicity against malignant mesothelioma, Am J Respir Crit Care Med, vol.181, pp.1383-90, 2010.

Y. P. Hung, F. Dong, J. C. Watkins, V. Nardi, R. Bueno et al., Identification of ALK rearrangements in malignant peritoneal mesothelioma, JAMA Oncol, vol.4, pp.235-243, 2018.

N. M. Joseph, Y. Y. Chen, A. Nasr, I. Yeh, E. Talevich et al., Genomic profiling of malignant peritoneal mesothelioma reveals recurrent alterations in epigenetic regulatory genes BAP1, SETD2, and DDX3X, Mod Pathol, vol.30, pp.246-54, 2017.

R. Shrestha, N. Nabavi, Y. Y. Lin, F. Mo, S. Anderson et al., BAP1 haploinsufficiency predicts a distinct immunogenic class of malignant peritoneal mesothelioma, Genome Med, 2019.

R. Bueno, E. W. Stawiski, L. D. Goldstein, S. Durinck, D. Rienzo et al., Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations, Nat Genet, vol.48, pp.407-423, 2016.

D. Jean, J. Daubriac, L. Pimpec-barthes, F. Galateau-salle, F. Jaurand et al., High-density array-CGH with targeted NGS unmask multiple noncontiguous minute deletions on chromosome 3p21 in mesothelioma, Arch Pathol Lab Med, vol.136, pp.13432-13439, 2012.

A. S. Mansfield, T. Peikert, J. B. Smadbeck, J. Udell, E. Garcia-rivera et al., Neoantigenic potential of complex chromosomal rearrangements in mesothelioma, J Thorac Oncol, vol.14, pp.276-87, 2019.

J. Hmeljak, F. Sanchez-vega, K. A. Hoadley, J. Shih, C. Stewart et al., Integrative molecular characterization of malignant pleural mesothelioma, Cancer Discov, vol.8, pp.1548-65, 2018.

A. Tallet, J. C. Nault, A. Renier, I. Hysi, F. Galateau-salle et al., Overexpression and promoter mutation of the TERT gene in malignant pleural mesothelioma, Oncogene, vol.33, pp.3748-52, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02478602

M. Betti, E. Casalone, D. Ferrante, A. Aspesi, G. Morleo et al., Germline mutations in DNA repair genes predispose asbestos-exposed patients to malignant pleural mesothelioma, Cancer Lett, vol.405, pp.38-45, 2017.

R. Hassan, B. Morrow, A. Thomas, T. Walsh, M. K. Lee et al., Inherited predisposition to malignant mesothelioma and overall survival following platinum chemotherapy, Proc Natl Acad Sci, vol.116, pp.9008-9021, 2019.

V. Panou, M. Gadiraju, A. Wolin, C. M. Weipert, E. Skarda et al., Frequency of germline mutations in cancer susceptibility genes in malignant mesothelioma, J Clin Oncol, vol.36, pp.2863-71, 2018.

S. Pastorino, Y. Yoshikawa, H. I. Pass, M. Emi, M. Nasu et al., A subset of mesotheliomas with improved survival occurring in carriers of BAP1 and other germline mutations, J Clin Oncol, vol.36, pp.3485-94, 2018.

J. R. Testa, M. Cheung, P. J. Below, J. E. Tan, Y. Sementino et al., Germline BAP1 mutations predispose to malignant mesothelioma, Nat Genet, vol.43, pp.1022-1027, 2011.

B. C. Christensen, E. A. Houseman, J. J. Godleski, C. J. Marsit, J. L. Longacker et al., Epigenetic profiles distinguish pleural mesothelioma from normal pleura and predict lung asbestos burden and clinical outcome, Cancer Res, vol.69, pp.227-261, 2009.

Y. Goto, K. Shinjo, Y. Kondo, L. Shen, M. Toyota et al., Epigenetic profiles distinguish malignant pleural mesothelioma from lung adenocarcinoma, Cancer Res, vol.69, pp.9073-82, 2009.

K. C. Mcloughlin, A. S. Kaufman, and D. S. Schrump, Targeting the epigenome in malignant pleural mesothelioma, Transl Lung Cancer Res, vol.6, pp.350-65, 2017.

L. M. Lafave, W. Beguelin, R. Koche, M. Teater, B. Spitzer et al., Loss of BAP1 function leads to EZH2-dependent transformation, Nat Med, vol.21, pp.1344-1353, 2015.

E. Felley-bosco and H. Rehrauer, Non-coding transcript heterogeneity in mesothelioma: insights from asbestos-exposed mice, Int J Mol Sci, vol.19, p.1163, 2018.

G. Reid, MicroRNAs in mesothelioma: from tumour suppressors and biomarkers to therapeutic targets, J Thorac Dis, vol.7, pp.1031-1071, 2015.

A. S. Singh, R. Heery, and S. G. Gray, In silico and in vitro analyses of LncRNAs as potential regulators in the transition from the epithelioid to sarcomatoid histotype of malignant pleural mesothelioma (MPM), Int J Mol Sci, vol.19, p.1297, 2018.

M. C. Jaurand and D. Jean, Biomolecular pathways and malignant pleural mesothelioma, Malignant Pleural Mesothelioma: Present Status and Future Directions, pp.169-92, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02479956

T. A. Yap, J. G. Aerts, S. Popat, and D. A. Fennell, Novel insights into mesothelioma biology and implications for therapy, Nat Rev Cancer, vol.17, pp.475-88, 2017.

A. N. Husain, T. Colby, N. Ordonez, T. Krausz, R. Attanoos et al., Guidelines for pathologic diagnosis of malignant mesothelioma: 2012 update of the consensus statement from the international mesothelioma interest group, Arch Pathol Lab Med, vol.137, pp.647-67, 2013.

K. Oehl, B. Vrugt, I. Opitz, and M. Meerang, Heterogeneity in Malignant Pleural Mesothelioma, Int J Mol Sci, p.19, 2018.

A. De-reynies, M. C. Jaurand, A. Renier, G. Couchy, I. Hysi et al., Molecular classification of malignant pleural mesothelioma: identification of a poor prognosis subgroup linked to the epithelialto-mesenchymal transition, Clin Cancer Res, vol.20, pp.1323-1357, 2014.

R. Tranchant, L. Quetel, A. Tallet, C. Meiller, A. Renier et al., Co-occurring mutations of tumor suppressor genes, LATS2 and NF2, in malignant pleural mesothelioma, Clin Cancer Res, vol.23, pp.3191-202, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-02478604

Y. Blum, C. Meiller, L. Quetel, N. Elarouci, M. Ayadi et al., Dissecting heterogeneity in malignant pleural mesothelioma through histomolecular gradients for clinical applications, Nat Commun, vol.10, p.1333, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02090279

Y. Blum, M. C. Jaurand, D. Reynies, A. Jean, and D. , Unraveling the cellular heterogeneity of malignant pleural mesothelioma through a deconvolution approach, Mol Cell Oncol, vol.6, p.1610322, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02478611

N. Alcala, L. Mangiante, N. Le-stang, C. E. Gustafson, S. Boyault et al., Redefining malignant pleural mesothelioma types as a continuum uncovers immune-vascular interactions, EBioMedicine, vol.48, pp.191-202, 2019.

S. Comertpay, S. Pastorino, M. Tanji, R. Mezzapelle, O. Strianese et al., Evaluation of clonal origin of malignant mesothelioma, J Transl Med, vol.12, p.301, 2014.

H. Oey, M. Daniels, V. Relan, T. M. Chee, M. R. Davidson et al., Wholegenome sequencing of human malignant mesothelioma tumours and cell lines, Carcinogenesis, vol.40, pp.724-758, 2019.

J. Minnema-luiting, H. Vroman, J. Aerts, and R. Cornelissen, Heterogeneity in immune cell content in malignant pleural mesothelioma, Int J Mol Sci, vol.19, p.1041, 2018.

I. C. Salaroglio, J. Kopecka, F. Napoli, M. Pradotto, F. Maletta et al., Potential diagnostic and prognostic role of microenvironment in malignant pleural mesothelioma, J Thorac Oncol, vol.14, pp.1458-71, 2019.

K. Kiyotani, J. H. Park, H. Inoue, A. Husain, S. Olugbile et al., Integrated analysis of somatic mutations and immune microenvironment in malignant pleural mesothelioma, Oncoimmunology, vol.6, p.1278330, 2017.

C. C. Maley, A. Aktipis, T. A. Graham, A. Sottoriva, A. M. Boddy et al., Classifying the evolutionary and ecological features of neoplasms, Nat Rev Cancer, vol.17, pp.605-624, 2017.

L. Mutti, T. Peikert, B. Robinson, A. Scherpereel, A. S. Tsao et al., Scientific advances and new frontiers in mesothelioma therapeutics, J Thorac Oncol, vol.13, pp.1269-83, 2018.

M. Tagawa, Y. Tada, H. Shimada, and K. Hiroshima, Gene therapy for malignant mesothelioma: current prospects and challenges, Cancer Gene Ther, vol.20, pp.150-156, 2013.

A. Lila, A. S. Kato, C. Fukushima, M. Huang, C. L. Wada et al., Downregulation of thymidylate synthase by RNAi molecules enhances the antitumor effect of pemetrexed in an orthotopic malignant mesothelioma xenograft mouse model, Int J Oncol, vol.48, pp.1399-407, 2016.

R. E. Favoni, A. Daga, P. Malatesta, and T. Florio, Preclinical studies identify novel targeted pharmacological strategies for treatment of human malignant pleural mesothelioma, Br J Pharmacol, vol.166, pp.532-53, 2012.

E. Giovannetti, P. A. Zucali, Y. G. Assaraf, L. G. Leon, K. Smid et al., Preclinical emergence of vandetanib as a potent antitumour agent in mesothelioma: molecular mechanisms underlying its synergistic interaction with pemetrexed and carboplatin, Br J Cancer, vol.105, pp.1542-53, 2011.

A. R. Hanauske, The role of alimta in the treatment of malignant pleural mesothelioma: an overview of preclinical and clinical trials

, Lung Cancer, pp.121-125, 2004.

K. Iwahori, S. Serada, M. Fujimoto, B. Ripley, S. Nomura et al., SOCS-1 gene delivery cooperates with cisplatin plus pemetrexed to exhibit preclinical antitumor activity against malignant pleural mesothelioma, Int J Cancer, vol.132, pp.459-71, 2013.

L. G. Leon, M. Gemelli, R. Sciarrillo, A. Avan, N. Funel et al., Synergistic activity of the c-Met and tubulin inhibitor tivantinib (ARQ197) with pemetrexed in mesothelioma cells, Curr Drug Targets, vol.15, pp.1331-1371, 2014.

T. Ueno, S. Toyooka, T. Fukazawa, T. Kubo, J. Soh et al., Preclinical evaluation of microRNA-34b/c delivery for malignant pleural mesothelioma, Acta Med Okayama, vol.68, pp.23-29, 2014.

H. Ando, S. Kobayashi, A. Lila, A. S. Eldin, N. E. Kato et al., Advanced therapeutic approach for the treatment of malignant pleural mesothelioma via the intrapleural administration of liposomal pemetrexed, J Control Release, vol.220, pp.29-36, 2015.

M. Meerang, A. Boss, D. Kenkel, A. Broggini-tenzer, K. Berard et al., Evaluation of imaging techniques for the assessment of tumour progression in an orthotopic rat model of malignant pleural mesotheliomadagger, Eur J Cardiothorac Surg, vol.47, pp.34-41, 2015.

S. Iochmann, S. Lerondel, C. Blechet, M. Lavergne, S. Pesnel et al., Monitoring of tumour progression using bioluminescence imaging and computed tomography scanning in a nude mouse orthotopic model of human small cell lung cancer, Lung Cancer, vol.77, pp.70-76, 2012.

T. K. Nayak, M. Bernardo, D. E. Milenic, P. L. Choyke, and M. W. Brechbiel, Orthotopic pleural mesothelioma in mice: SPECT/CT and MR imaging with HER1-and HER2-targeted radiolabeled antibodies, Radiology, vol.267, pp.173-82, 2013.

A. M. Behbehani, W. J. Hunter, A. L. Chapman, and F. Lin, Studies of a human mesothelioma, Hum Pathol, vol.13, pp.862-868, 1982.

F. R. Reale, T. W. Griffin, J. M. Compton, S. Graham, P. L. Townes et al., Characterization of a human malignant mesothelioma cell line (H-MESO-1): a biphasic solid and ascitic tumor model, Cancer Res, vol.47, pp.3199-205, 1987.

F. Gueugnon, S. Leclercq, C. Blanquart, C. Sagan, L. Cellerin et al., Identification of novel markers for the diagnosis of malignant pleural mesothelioma, Am J Pathol, vol.178, pp.1033-1075, 2011.

D. Jean, E. Thomas, M. E. Renier, A. De-reynies, A. Lecomte et al., Syntenic relationships between genomic profiles of fiber-induced murine and human malignant mesothelioma, Am J Pathol, vol.178, pp.881-94, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00531343

R. Tranchant, L. Quetel, F. Montagne, D. Wolf, J. Meiller et al., Assessment of signaling pathway inhibitors and identification of predictive biomarkers in malignant pleural mesothelioma, Lung Cancer, vol.126, pp.15-24, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02478609

L. Zeng, A. Buard, I. Monnet, C. Boutin, J. Fleury et al., In vitro effects of recombinant human interferon gamma on human mesothelioma cell lines, Int J Cancer, vol.55, pp.515-535, 1993.

L. Zeng, J. Fleury-feith, I. Monnet, C. Boutin, J. Bignon et al., Immunocytochemical characterization of cell lines from human malignant mesothelioma: characterization of human mesothelioma cell lines by immunocytochemistry with a panel of monoclonal antibodies, Hum Pathol, vol.25, pp.227-261, 1994.

T. Chernova, X. M. Sun, I. R. Powley, S. Galavotti, S. Grosso et al., Molecular profiling reveals primary mesothelioma cell lines recapitulate human disease, Cell Death Differ, vol.23, pp.1152-64, 2016.

P. Smeele, S. M. Almeida, C. Meiller, A. L. Chene, C. Liddell et al., Brain-derived neurotrophic factor, a new soluble biomarker for malignant pleural mesothelioma involved in angiogenesis, Mol Cancer, vol.17, p.148, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01904612

J. Jongsma, E. Van-montfort, M. Vooijs, J. Zevenhoven, P. Krimpenfort et al., A conditional mouse model for malignant mesothelioma, Cancer Cell, vol.13, pp.261-71, 2008.

D. Jean and M. C. Jaurand, Mesotheliomas in Genetically engineered mice unravel mechanism of mesothelial carcinogenesis, Int J Mol Sci, vol.19, p.2191, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02478607

A. M. Kukuyan, E. Sementino, Y. Kadariya, C. W. Menges, M. Cheung et al., Inactivation of Bap1 cooperates with losses of Nf2 and Cdkn2a to drive the development of pleural malignant mesothelioma in conditional mouse models, Cancer Res, vol.79, pp.4113-4136, 2019.

B. A. Ruggeri, F. Camp, and S. Miknyoczki, Animal models of disease: preclinical animal models of cancer and their applications and utility in drug discovery, Biochem Pharmacol, vol.87, pp.150-61, 2014.

M. Singh, C. L. Murriel, and L. Johnson, Genetically engineered mouse models: closing the gap between preclinical data and trial outcomes, Cancer Res, vol.72, pp.2695-700, 2012.

M. J. Disselhorst, J. Quispel-janssen, F. Lalezari, K. Monkhorst, J. F. De-vries et al., Ipilimumab and nivolumab in the treatment of recurrent malignant pleural mesothelioma (INITIATE): results of a prospective, single-arm, phase 2 trial, Lancet Respir Med, vol.7, issue.18, p.30420, 2019.

D. A. Fennell, E. Kirkpatrick, K. Cozens, M. Nye, J. Lester et al., CONFIRM: a double-blind, placebo-controlled phase III clinical trial investigating the effect of nivolumab in patients with relapsed mesothelioma: study protocol for a randomised controlled trial, Trials, vol.19, p.233, 2018.

A. S. Mansfield and M. G. Zauderer, Nivo-lution in mesothelioma, Clin Cancer Res, vol.25, pp.5438-5478, 2019.

P. A. Moalli, J. L. Macdonald, L. A. Goodglick, and A. B. Kane, Acute injury and regeneration of the mesothelium in response to asbestos fibers, Am J Pathol, vol.128, pp.426-471, 1987.

C. Jackaman, C. S. Bundell, and B. F. Kinnear,

D. Hagen, IL-2 intratumoral immunotherapy enhances CD8+ T cells that mediate destruction of tumor cells and tumor-associated vasculature: a novel mechanism for IL-2, J Immunol, vol.171, pp.5051-63, 2003.

M. R. Davis, L. S. Manning, D. Whitaker, M. J. Garlepp, and B. W. Robinson, Establishment of a murine model of malignant mesothelioma, Int J Cancer, vol.52, pp.881-887, 1992.

J. E. Craighead, N. J. Akley, L. B. Gould, and B. L. Libbus, Characteristics of tumors and tumor cells cultured from experimental asbestos-induced mesotheliomas in rats, Am J Pathol, vol.129, pp.448-62, 1987.

D. Roulois, S. Deshayes, M. N. Guilly, J. S. Nader, C. Liddell et al., Characterization of preneoplastic and neoplastic rat mesothelial cell lines: the involvement of TETs, DNMTs, and 5-hydroxymethylcytosine, Oncotarget, vol.7, pp.34664-87, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01306838

J. S. Nader, J. Abadie, S. Deshayes, A. Boissard, S. Blandin et al., Characterization of increasing stages of invasiveness identifies stromal/cancer cell crosstalk in rat models of mesothelioma, Oncotarget, vol.9, pp.16311-16340, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01726393

Y. Shi, A. Hollenstein, E. Felley-bosco, C. Fraefel, M. Ackermann et al., Bioluminescence imaging for in vivo monitoring of local recurrence mesothelioma model, Lung Cancer, vol.71, pp.370-371, 2011.

M. C. Crisanti, A. F. Wallace, V. Kapoor, F. Vandermeers, M. L. Dowling et al., The HDAC inhibitor panobinostat (LBH589) inhibits mesothelioma and lung cancer cells in vitro and in vivo with particular efficacy for small cell lung cancer, Mol Cancer Ther, vol.8, pp.2221-2252, 2009.

V. Laszlo, Z. Valko, J. Ozsvar, I. Kovacs, T. Garay et al., The FAK inhibitor BI 853520 inhibits spheroid formation and orthotopic tumor growth in malignant pleural mesothelioma, J Mol Med, vol.97, pp.231-273, 2019.

C. Pulito, E. Korita, A. Sacconi, M. Valerio, L. Casadei et al., Dropwort-induced metabolic reprogramming restrains YAP/TAZ/TEAD oncogenic axis in mesothelioma, J Exp Clin Cancer Res, vol.38, p.349, 2019.

S. Abe, M. K. Kaneko, Y. Tsuchihashi, T. Izumi, S. Ogasawara et al., Antitumor effect of novel anti-podoplanin antibody NZ-12 against malignant pleural mesothelioma in an orthotopic xenograft model, Cancer Sci, vol.107, pp.1198-205, 2016.

S. Golfier, C. Kopitz, A. Kahnert, I. Heisler, C. A. Schatz et al., Anetumab ravtansine: a novel mesothelin-targeting antibodydrug conjugate cures tumors with heterogeneous target expression favored by bystander effect, Mol Cancer Ther, vol.13, pp.1537-1585, 2014.

H. Krishnan, J. Rayes, T. Miyashita, G. Ishii, E. P. Retzbach et al., Podoplanin: an emerging cancer biomarker and therapeutic target, Cancer Sci, vol.109, pp.1292-1301, 2018.

S. J. Scales, N. Gupta, G. Pacheco, R. Firestein, D. M. French et al., An antimesothelin-monomethyl auristatin e conjugate with potent antitumor activity in ovarian, pancreatic, and mesothelioma models, Mol Cancer Ther, vol.13, pp.2630-2670, 2014.

Y. F. Zhang, Y. Phung, W. Gao, S. Kawa, R. Hassan et al., New high affinity monoclonal antibodies recognize non-overlapping epitopes on mesothelin for monitoring and treating mesothelioma, Sci Rep, vol.5, p.9928, 2015.

J. Zhang, S. Khanna, Q. Jiang, C. Alewine, M. Miettinen et al., Efficacy of anti-mesothelin immunotoxin RG7787 plus nab-paclitaxel against mesothelioma patient-derived xenografts and mesothelin as a biomarker of tumor response, Clin Cancer Res, vol.23, pp.1564-74, 2017.

E. K. Moon, C. Carpenito, J. Sun, L. C. Wang, V. Kapoor et al., Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelinspecific chimeric antibody receptor, Clin Cancer Res, vol.17, pp.4719-4749, 2011.

Y. Zhao, E. Moon, C. Carpenito, C. M. Paulos, X. Liu et al., Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor, Cancer Res, vol.70, pp.9053-61, 2010.

J. Jung, H. S. Seol, and S. Chang, The generation and application of patient-derived xenograft model for cancer research, Cancer Res Treat, vol.50, pp.1-10, 2018.

A. P. Chahinian, J. T. Beranek, Y. Suzuki, J. G. Bekesi, L. Wisniewski et al., Transplantation of human malignant mesothelioma into nude mice, Cancer Res, vol.40, pp.181-186, 1980.

L. Wu, G. Allo, T. John, M. Li, T. Tagawa et al., Patient-derived xenograft establishment from human malignant pleural mesothelioma, Clin Cancer Res, vol.23, pp.1060-1067, 2017.

X. Cao, E. W. Shores, J. Hu-li, M. R. Anver, B. L. Kelsall et al., Defective lymphoid development in mice lacking expression of the common cytokine receptor gamma chain, Immunity, vol.2, pp.223-261, 1995.

L. B. Weiswald and D. Bellet, Dangles-Marie V. Spherical cancer models in tumor biology, Neoplasia, vol.17, pp.1-15, 2015.

H. Kim, Y. Phung, and M. Ho, Changes in global gene expression associated with 3D structure of tumors: an ex vivo matrix-free mesothelioma spheroid model, PLoS ONE, vol.7, p.39556, 2012.

D. Barbone, L. Van-dam, C. Follo, P. V. Jithesh, S. D. Zhang et al., Analysis of gene expression in 3D spheroids highlights a survival role for ASS1 in mesothelioma, PLoS ONE, vol.11, 2016.

D. Barbone, T. M. Yang, J. R. Morgan, G. Gaudino, and V. C. Broaddus, Mammalian target of rapamycin contributes to the acquired apoptotic resistance of human mesothelioma multicellular spheroids, J Biol Chem, vol.283, pp.13021-13051, 2008.

D. Barbone, P. Cheung, S. Battula, S. Busacca, S. G. Gray et al., Vorinostat eliminates multicellular resistance of mesothelioma 3D spheroids via restoration of noxa expression, PLoS ONE, vol.7, p.52753, 2012.

J. Daubriac, J. Fleury-feith, L. Kheuang, J. Galipon, A. Saint-albin et al., Malignant pleural mesothelioma cells resist anoikis as quiescent pluricellular aggregates, Cell Death Differ, vol.16, pp.1146-55, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00388174

I. Gerogianni, E. Pitaraki, R. M. Jagirdar, O. Kouliou, L. Giannakou et al., 2-Deoxy-glucose enhances the effect of cisplatin and pemetrexed in reducing malignant pleural mesothelioma cell proliferation but not spheroid growth, Anticancer Res, vol.39, pp.3809-3823, 2019.

H. Lei, S. C. Hofferberth, R. Liu, A. Colby, K. M. Tevis et al., Paclitaxelloaded expansile nanoparticles enhance chemotherapeutic drug delivery in mesothelioma 3-dimensional multicellular spheroids, J Thorac Cardiovasc Surg, vol.149, pp.1417-1441, 2015.

C. Linot, J. Poly, J. Boucard, D. Pouliquen, S. Nedellec et al., PEGylated anionic magnetofluorescent nanoassemblies: impact of their interface structure on magnetic resonance imaging contrast and cellular uptake, ACS Appl Mater Interfaces, vol.9, pp.14242-57, 2017.

Y. T. Phung, D. Barbone, V. C. Broaddus, and M. Ho, Rapid generation of in vitro multicellular spheroids for the study of monoclonal antibody therapy, J Cancer, vol.2, pp.507-521, 2011.

X. Xiang, Y. Phung, M. Feng, K. Nagashima, J. Zhang et al., The development and characterization of a human mesothelioma in vitro 3D model to investigate immunotoxin therapy, PLoS ONE, vol.6, 2011.

D. Barbone, J. A. Ryan, N. Kolhatkar, A. D. Chacko, D. M. Jablons et al., The Bcl-2 repertoire of mesothelioma spheroids underlies acquired apoptotic multicellular resistance, Cell Death Dis, vol.2, 2011.

N. Echeverry, G. Ziltener, D. Barbone, W. Weder, R. A. Stahel et al., Inhibition of autophagy sensitizes malignant pleural mesothelioma cells to dual PI3K/mTOR inhibitors, Cell Death Dis, vol.6, p.1757, 2015.

C. Follo, D. Barbone, W. G. Richards, R. Bueno, and V. C. Broaddus, Autophagy initiation correlates with the autophagic flux in 3D models of mesothelioma and with patient outcome, Autophagy, vol.12, pp.1180-94, 2016.

K. U. Kim, S. M. Wilson, K. S. Abayasiriwardana, R. Collins, L. Fjellbirkeland et al., A novel in vitro model of human mesothelioma for studying tumor biology and apoptotic resistance, Am J Respir Cell Mol Biol, vol.33, pp.541-549, 2005.

J. Kuen, D. Darowski, T. Kluge, and M. Majety, Pancreatic cancer cell/fibroblast co-culture induces M2 like macrophages that influence therapeutic response in a 3D model, PLoS ONE, vol.12, p.182039, 2017.

K. M. Tevis, R. J. Cecchi, Y. L. Colson, and M. W. Grinstaff, Mimicking the tumor microenvironment to regulate macrophage phenotype and assessing chemotherapeutic efficacy in embedded cancer cell/macrophage spheroid models, Acta Biomater, vol.50, pp.271-280, 2017.

S. P. Rebelo, C. Pinto, T. R. Martins, N. Harrer, M. F. Estrada et al., 3D-3-culture: a tool to unveil macrophage plasticity in the tumour microenvironment, Biomaterials, vol.163, pp.185-97, 2018.

J. S. De-bono and A. Ashworth, Translating cancer research into targeted therapeutics, Nature, vol.467, pp.543-552, 2010.

L. Moreno and A. D. Pearson, How can attrition rates be reduced in cancer drug discovery?, Exp Opin Drug Discov, vol.8, pp.363-371, 2013.

M. L. Graham and M. J. Prescott, The multifactorial role of the 3Rs in shifting the harm-benefit analysis in animal models of disease, Eur J Pharmacol, vol.759, pp.19-29, 2015.

C. Jean-quartier, F. Jeanquartier, I. Jurisica, and A. Holzinger, In silico cancer research towards 3R, BMC Cancer, vol.18, p.408, 2018.

W. Sun, Z. Luo, J. Lee, H. J. Kim, K. Lee et al., Organ-on-a-chip for cancer and immune organs modeling, Adv Healthc Mater, vol.8, p.1801363, 2019.

C. G. Begley and L. M. Ellis, Drug development: raise standards for preclinical cancer research, Nature, vol.483, pp.531-534, 2012.

J. Kimmelman and C. Federico, Consider drug efficacy before first-in-human trials, Nature, vol.542, pp.25-32, 2017.