A. Boyer, E. Pasquier, P. Tomasini, J. Ciccolini, L. Greillier et al., Drug repurposing in malignant pleural mesothelioma: A breath of fresh air?, Eur. Respir. Rev, vol.27, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02143583

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.

A. Singh, N. Pruett, and C. D. Hoang, In vitro experimental models of mesothelioma revisited, Transl. Lung Cancer Res, vol.6, pp.248-258, 2017.

D. Katzman and D. H. Sterman, Updates in the diagnosis and treatment of malignant pleural mesothelioma, Curr. Opin. Pulm. Med, vol.24, pp.319-326, 2018.

A. Lacourt, E. Leveque, M. Goldberg, and K. Leffondre, Dose-time response association between occupational asbestos exposure and pleural mesothelioma: Authors' response, Occup. Environ. Med, vol.75, pp.161-162, 2018.

A. J. Mccambridge, A. Napolitano, A. S. Mansfield, D. A. Fennell, Y. Sekido et al., Progress in the Management of Malignant Pleural Mesothelioma in 2017, J. Thorac. Oncol, vol.13, pp.606-623, 2018.

G. Serio, V. Pagliarule, A. Marzullo, F. Pezzuto, M. Gentile et al., Molecular changes of malignant mesothelioma in the testis and their impact on prognosis: Analyses of two cases, Int. J. Clin. Exp. Pathol, vol.9, pp.7658-7667, 2016.

E. Goswami, V. Craven, D. L. Dahlstrom, D. Alexander, and F. Mowat, Domestic asbestos exposure: A review of epidemiologic and exposure data, Int. J. Environ. Res. Public Health, vol.10, pp.5629-5670, 2013.

C. W. Noonan, Environmental asbestos exposure and risk of mesothelioma, Ann. Transl. Med, 2017.

G. Serio, F. Pezzuto, A. Marzullo, A. Scattone, D. Cavone et al., Report of a Case with Long Survival (Seventeen Years) Analyzed by Cgh-Array, Peritoneal Mesothelioma with Residential Asbestos Exposure, vol.18, 1818.

L. Vimercati, D. Cavone, P. Lovreglio, L. De-maria, A. Caputi et al., Environmental asbestos exposure and mesothelioma cases in Bari, Apulia region, southern Italy: A national interest site for land reclamation, Environ. Sci. Pollut, vol.25, pp.15692-15701, 2018.

A. N. Husain, T. V. Colby, N. G. Ordonez, T. C. Allen, R. L. Attanoos et al., Guidelines for Pathologic Diagnosis of Malignant Mesothelioma 2017 Update of the Consensus Statement From the International Mesothelioma Interest Group, Arch. Pathol. Lab. Med, vol.142, pp.89-108, 2018.

S. E. Mutsaers, M. C. Jaurand, Y. C. Garylee, and C. M. Prele, Mesothelial cells and pleural immunology, Textbook of Pleural Diseases, pp.27-44, 2016.

N. S. Wang, Anatomy of the pleura, Clin. Chest Med, vol.19, pp.229-240, 1998.

H. Oshiro, M. Miura, H. Iobe, T. Kudo, Y. Shimazu et al., Lymphatic Stomata in the Adult Human Pulmonary Ligament, Lymphat. Res. Biol, vol.13, pp.137-145, 2015.

V. C. Broaddus, J. I. Everitt, B. Black, and A. B. Kane, Non-neoplastic and neoplastic pleural endpoints following fiber exposure, J. Toxicol. Environ. Health B Crit. Rev, vol.14, pp.153-178, 2011.

J. Fleury-feith and M. C. Jaurand, Pleural lymphatics and pleural diseases related to fibres, Rev. Pneumol. Clin, vol.69, pp.358-362, 2013.

K. Oehl, B. Vrugt, I. Opitz, and M. Meerang, Heterogeneity in Malignant Pleural Mesothelioma, Int. J. Mol. Sci, vol.19, 1603.

M. Hylebos, G. Van-camp, G. Vandeweyer, E. Fransen, M. Beyens et al., Large-scale copy number analysis reveals variations in genes not previously associated with malignant pleural mesothelioma, Oncotarget, vol.8, pp.113673-113686, 2017.

D. Jean, J. Daubriac, F. Le-pimpec-barthes, F. Galateau-salle, and M. C. Jaurand, Molecular changes in mesothelioma with an impact on prognosis and treatment, Arch. Pathol. Lab. Med, vol.136, pp.277-293, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-02478588

R. Tranchant, L. Quetel, A. Tallet, C. Meiller, A. Renier et al., Co-occurring Mutations of Tumor Suppressor Genes, LATS2 and NF2, Malignant Pleural Mesothelioma. Clin. Cancer Res, vol.23, pp.3191-3202, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-02478604

R. Bueno, E. W. Stawiski, L. D. Goldstein, S. Durinck, A. De-rienzo et al., Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations, Nat. Genet, vol.48, pp.407-416, 2016.

D. Jean, E. Thomas, A. Renier, A. De-reynies, C. Lecomte et al., Syntenic relationships between genomic profiles of fiber-induced murine and human malignant mesothelioma, Am. J. Pathol, vol.176, pp.881-894, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00531343

A. C. Borczuk, J. Pei, R. N. Taub, B. Levy, O. Nahum et al., Genome-wide analysis of abdominal and pleural malignant mesothelioma with DNA arrays reveals both common and distinct regions of copy number alteration, Cancer Biol. Ther, vol.17, pp.328-335, 2016.

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, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02478602

E. Barone, F. Gemignani, and S. Landi, Overexpressed genes in malignant pleural mesothelioma: Implications in clinical management, J. Thorac. Dis, vol.10, pp.369-382, 2018.

E. Riquelme, M. B. Suraokar, J. Rodriguez, B. Mino, H. Y. Lin et al., Wistuba, I.I. Frequent coamplification and cooperation between C-MYC and PVT1 oncogenes promote malignant pleural mesothelioma, J. Thorac. Oncol, vol.9, pp.998-1007, 2014.

A. S. Tsao, N. Harun, J. Fujimoto, V. Devito, J. J. Lee et al., Elevated PDGFRB gene copy number gain is prognostic for improved survival outcomes in resected malignant pleural mesothelioma, Ann. Diagn. Pathol, vol.18, pp.140-145, 2014.

L. Quinn, S. P. Finn, S. Cuffe, and S. G. Gray, Non-coding RNA repertoires in malignant pleural mesothelioma, Lung Cancer, vol.90, pp.417-426, 2015.

I. Vannini, F. Fanini, and M. Fabbri, Emerging roles of microRNAs in cancer, Curr. Opin. Genet. Dev, vol.48, pp.128-133, 2018.

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.227-234, 2009.

B. C. Christensen, J. J. Godleski, C. J. Marsit, E. A. Houseman, C. Y. Lopez-fagundo et al., Asbestos exposure predicts cell cycle control gene promoter methylation in pleural mesothelioma, Carcinogenesis, vol.29, pp.1555-1559, 2008.

R. Bruno, G. Ali, R. Giannini, A. Proietti, M. Lucchi et al., Malignant pleural mesothelioma and mesothelial hyperplasia: A new molecular tool for the differential diagnosis, Oncotarget, vol.8, pp.2758-2770, 2017.

S. Busacca, S. Germano, L. De-cecco, M. Rinaldi, F. Comoglio et al., MicroRNA signature of malignant mesothelioma with potential diagnostic and prognostic implications, Am. J. Respir. Cell Mol. Biol, vol.42, pp.312-319, 2010.

G. Reid, MicroRNAs in mesothelioma: From tumour suppressors and biomarkers to therapeutic targets, J. Thorac. Dis, vol.7, pp.1031-1040, 2015.

D. Santi, C. Melaiu, O. Bonotti, A. Cascione, L. Di-leva et al., Deregulation of miRNAs in malignant pleural mesothelioma is associated with prognosis and suggests an alteration of cell metabolism, vol.7, p.3140, 2017.

M. C. Jaurand and D. Jean, Biomolecular Pathways and Malignant Pleural Mesothelioma, Malignant Pleural Mesothelioma: Present Status and Future Directions, pp.173-196, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02479956

Y. Sekido, Molecular pathogenesis of malignant mesothelioma, Carcinogenesis, vol.34, pp.1413-1419, 2013.

E. Felley-bosco, I. Opitz, and M. Meerang, Hedgehog Signaling in Malignant Pleural Mesothelioma, Genes, vol.6, pp.500-511, 2015.

A. C. Gradilla, D. Sanchez-hernandez, L. Brunt, and S. Scholpp, From top to bottom: Cell polarity in Hedgehog and Wnt trafficking, BMC Biol, vol.16, 2018.

E. G. Leprieur, D. M. Jablons, and B. He, Old Sonic Hedgehog, new tricks: A new paradigm in thoracic malignancies, Oncotarget, vol.9, pp.14680-14691, 2018.

M. B. Suraokar, M. I. Nunez, L. Diao, C. W. Chow, D. Kim et al., Expression profiling stratifies mesothelioma tumors and signifies deregulation of spindle checkpoint pathway and microtubule network with therapeutic implications, Ann. Oncol, vol.25, pp.1184-1192, 2014.

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 epithelial-to-mesenchymal transition, Clin. Cancer Res, vol.20, pp.1323-1334, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02478599

A. Churg, R. Attanoos, A. C. Borczuk, L. R. Chirieac, F. Galateau-salle et al., Dataset for Reporting of Malignant Mesothelioma of the Pleura or Peritoneum: Recommendations From the International Collaboration on Cancer Reporting (ICCR), Arch. Pathol. Lab. Med, vol.140, pp.1104-1110, 2016.

A. Bridda, I. Padoan, R. Mencarelli, and M. Frego, Peritoneal mesothelioma: A review, vol.9, 2007.

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.246-254, 2017.

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.235-238, 2018.

H. Tanigawa, H. Onodera, and A. Maekawa, Spontaneous Mesotheliomas in Fischer Rats-A Histological and Electron Microscopic Study, Toxicol. Pathol, vol.15, pp.157-163, 1987.

R. R. Maronpot, A. Nyska, J. E. Foreman, and Y. Ramot, The legacy of the F344 rat as a cancer bioassay model (a retrospective summary of three common F344 rat neoplasms), Crit. Rev. Toxicol, vol.46, pp.641-675, 2016.

E. Bomhard, Frequency of spontaneous tumours in NMRI mice in 21-month studies, Exp. Toxicol. Pathol, vol.45, pp.269-289, 1993.

R. L. Peters, L. S. Rabstein, G. J. Spahn, R. M. Madison, and R. J. Huebner, Incidence of spontaneous neoplasms in breeding and retired breeder BALB-cCr mice throughout the natural life span, Int. J. Cancer, vol.10, pp.273-282, 1972.

M. Shirai, T. Maejima, T. Tanimoto, K. Kumagai, T. Makino et al., Mixed Type of Malignant Mesothelioma in an Aged Male ICR Mouse, J. Toxicol. Pathol, vol.24, pp.169-172, 2011.

, Man-made vitreous fibres and Radon, IARC Monogr. Eval. Carcinog. Risks Hum, vol.43, pp.1-300, 1988.

G. Boulanger, P. Andujar, J. C. Pairon, M. A. Billon-galland, C. Dion et al., Quantification of short and long asbestos fibers to assess asbestos exposure: A review of fiber size toxicity, Environ. Health, vol.13, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01073918

J. C. Wagner and G. Berry, Mesotheliomas in rats following inoculation with asbestos, Br. J. Cancer, vol.23, pp.567-581, 1969.

S. Sneddon, A. M. Patch, I. M. Dick, S. Kazakoff, J. V. Pearson et al., Whole exome sequencing of an asbestos-induced wild-type murine model of malignant mesothelioma, BMC Cancer, vol.17, 2017.

T. Chernova, F. A. Murphy, S. Galavotti, X. M. Sun, I. R. Powley et al., Long-Fiber Carbon Nanotubes Replicate Asbestos-Induced Mesothelioma with Disruption of the Tumor Suppressor Gene Cdkn2a

, Curr. Biol, vol.27, pp.3302-3314, 2017.

Y. Guo, L. R. Chirieac, R. Bueno, H. Pass, W. Wu et al., Tsc1-Tp53 loss induces mesothelioma in mice, and evidence for this mechanism in human mesothelioma, Oncogene, vol.33, pp.3151-3160, 2014.

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.261-271, 2008.

Y. Kadariya, M. Cheung, J. Xu, J. Pei, E. Sementino et al., Bap1 Is a Bona Fide Tumor Suppressor: Genetic Evidence from Mouse Models Carrying Heterozygous Germline Bap1 Mutations, Cancer Res, vol.76, pp.2836-2844, 2016.

E. Sementino, C. W. Menges, Y. Kadariya, S. Peri, J. Xu et al.,

A. J. Klein-szanto, Inactivation of Tp53 and Pten drives rapid development of pleural and peritoneal malignant mesotheliomas, J. Cell. Physiol, 2018.

D. A. Altomare, C. W. Menges, J. Pei, L. Zhang, K. L. Skele-stump et al., Activated TNF-?/NF-?B signaling via down-regulation of Fas-associated factor 1 in asbestos-induced mesotheliomas from Arf knockout mice, Proc. Natl. Acad. Sci, vol.106, pp.3420-3425, 2009.

D. A. Altomare, C. W. Menges, J. Xu, J. Pei, L. Zhang et al., Losses of both products of the Cdkn2a/Arf locus contribute to asbestos-induced mesothelioma development and cooperate to accelerate tumorigenesis, PLoS ONE, vol.6, 2011.

D. A. Altomare, C. A. Vaslet, K. L. Skele, A. De-rienzo, K. Devarajan et al., A mouse model recapitulating molecular features of human mesothelioma, Cancer Res, vol.65, pp.8090-8095, 2005.

P. Andujar, C. Lecomte, A. Renier, J. Fleury-feith, L. Kheuang et al., Clinico-pathological features and somatic gene alterations in refractory ceramic fibre-induced murine mesothelioma reveal mineral fibre-induced mesothelioma identities, Carcinogenesis, vol.28, pp.1599-1605, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00329999

J. Fleury-feith, C. Lecomte, A. Renier, M. Matrat, L. Kheuang et al., Hemizygosity of N?2 is associated with increased susceptibility to asbestos-induced peritoneal tumours, Oncogene, vol.22, pp.3799-3805, 2003.

A. Napolitano, L. Pellegrini, A. Dey, D. Larson, M. Tanji et al., Minimal asbestos exposure in germline BAP1 heterozygous mice is associated with deregulated inflammatory response and increased risk of mesothelioma, Oncogene, vol.35, 1996.

C. A. Vaslet, N. J. Messier, and A. B. Kane, Accelerated progression of asbestos-induced mesotheliomas in heterozygous p53 +/? mice, Toxicol. Sci, vol.68, pp.331-338, 2002.

C. W. Menges, Y. Kadariya, D. Altomare, J. Talarchek, E. Neumann-domer et al., Tumor suppressor alterations cooperate to drive aggressive mesotheliomas with enriched cancer stem cells via a p53-miR-34a-c-Met axis, Cancer Res, vol.74, pp.1261-1271, 2014.

A. Takagi, A. Hirose, T. Nishimura, N. Fukumori, A. Ogata et al., Induction of mesothelioma in p53 +/? mouse by intraperitoneal application of multi-wall carbon nanotube, J. Toxicol. Sci, vol.33, pp.105-116, 2008.

Y. Kadariya, C. W. Menges, J. Talarchek, K. Q. Cai, A. J. Klein-szanto et al., Inflammation-Related IL1beta/IL1R Signaling Promotes the Development of Asbestos-Induced Malignant Mesothelioma, Cancer Prev. Res, vol.9, pp.406-414, 2016.

C. Robinson, I. Van-bruggen, A. Segal, M. Dunham, A. Sherwood et al., A novel SV40 TAg transgenic model of asbestos-induced mesothelioma: Malignant transformation is dose dependent, Cancer Res, vol.66, pp.10786-10794, 2006.

C. Robinson, I. M. Dick, M. J. Wise, A. Holloway, D. Diyagama et al., Consistent gene expression profiles in MexTAg transgenic mouse and wild type mouse asbestos-induced mesothelioma, BMC Cancer, vol.15, 2015.

C. Lecomte, P. Andujar, A. Renier, L. Kheuang, V. Abramowski et al., Similar tumor suppressor gene alteration profiles in asbestos-induced murine and human mesothelioma, Cell Cycle, vol.4, pp.1862-1869, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01928388

H. Rehrauer, L. Wu, W. Blum, L. Pecze, T. Henzi et al., How asbestos drives the tissue towards tumors: YAP activation, macrophage and mesothelial precursor recruitment, RNA editing, and somatic mutations, Oncogene, vol.37, pp.2645-2659, 2018.

J. Xu, Y. Kadariya, M. Cheung, J. Pei, J. Talarchek et al., Germline mutation of Bap1 accelerates development of asbestos-induced malignant mesothelioma, Cancer Res, vol.74, pp.4388-4397, 2014.

G. R. Bignell, C. D. Greenman, H. Davies, A. P. Butler, S. Edkins et al., Signatures of mutation and selection in the cancer genome, Nature, vol.463, pp.893-898, 2010.

P. Moller, P. H. Danielsen, K. Jantzen, M. Roursgaard, and S. Loft, Oxidatively damaged DNA in animals exposed to particles, Crit. Rev. Toxicol, vol.43, pp.96-118, 2013.