J. C. Wagner, C. A. Sleggs, and P. Marchand, Diffuse pleural mesothelioma and asbestos exposure in the North Western Cape Province, Br J Ind Med, vol.17, pp.260-71, 1960.

M. Albin, C. Magnani, S. Krstev, E. Rapiti, and I. Shefer, Asbestos and cancer: An overview of current trends in Europe, Environ Health Perspect, vol.107, issue.2, pp.289-98, 1999.

T. Kishimoto, S. Ozaki, K. Kato, H. Nishi, and K. Genba, Malignant pleural mesothelioma in parts of Japan in relationship to asbestos exposure, Ind Health, vol.42, issue.4, pp.435-444, 2004.

M. Goldberg, E. Imbernon, P. Rolland, G. Soit-ilg, A. Saves et al., The French National Mesothelioma Surveillance Program, Occup Environ Med, vol.63, issue.6, pp.390-395, 2006.

E. K. Park, K. M. Hannaford-turner, R. A. Hyland, A. R. Johnson, and D. H. Yates, Asbestos-related occupational lung diseases in NSW, Australia and potential exposure of the general population, Ind Health, vol.46, issue.6, pp.535-575, 2008.

R. L. Attanoos, A. Churg, F. Galateau-salle, A. R. Gibbs, and V. L. Roggli, Malignant Mesothelioma and Its Non-Asbestos Causes, Arch Pathol Lab Med, vol.142, issue.6, pp.753-60, 2018.

Y. Grosse, D. Loomis, K. Z. Guyton, B. Lauby-secretan, E. Ghissassi et al., Carcinogenicity of fluoro-edenite, silicon carbide fibres and whiskers, and carbon nanotubes, Lancet Oncol, vol.15, issue.13, pp.1427-1435, 2014.

A. B. Kane, R. H. Hurt, and H. Gao, The asbestos-carbon nanotube analogy: An update, Toxicol Appl Pharmacol, 2018.

K. Donaldson, C. A. Poland, F. A. Murphy, M. Macfarlane, T. Chernova et al., Pulmonary toxicity of carbon nanotubes and asbestos -Similarities and differences, Adv Drug Deliv Rev, vol.65, issue.15, pp.2078-86, 2013.

M. C. Jaurand, A. Renier, J. Daubriac, and . Mesothelioma, Do asbestos and carbon nanotubes pose the same health risk?, Part Fibre Toxicol, vol.6, p.16, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00387933

E. D. Kuempel, M. C. Jaurand, P. Moller, Y. Morimoto, N. Kobayashi et al., Evaluating the mechanistic evidence and key data gaps in assessing the potential carcinogenicity of carbon nanotubes and nanofibers in humans, Crit Rev Toxicol, vol.47, issue.1, pp.1-58, 2017.

A. Kane, D. Jean, S. Knuutila, and M. C. Jaurand, Malignant Mesothelioma: Mechanism of Carcinogenesis, Occupational Cancers, p.299319, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02479963

M. Lippmann, D. B. Yeates, and R. E. Albert, Deposition, retention and clearance of inhaled particles, Br J Industr Med, vol.37, pp.337-62, 1980.

, Man-made vitreous fibres2002

G. D. Nielsen and I. K. Koponen, Insulation fiber deposition in the airways of men and rats. A review of experimental and computational studies, Regul Toxicol Pharmacol, vol.94, pp.252-70, 2018.

B. Asgharian, T. P. Owen, E. D. Kuempel, and A. M. Jarabek, Dosimetry of inhaled elongate mineral particles in the respiratory tract: The impact of shape factor, Toxicol Appl Pharmacol, 2018.

G. Oberdorster, Evaluation and use of animal models to assess mechanisms of fibre carcinogenicity, IARC Sci Publ, issue.140, pp.107-132, 1996.

G. A. Miserocchi, G. A. Sancini, F. Mantegazza, and G. Chiappino, Translocation pathways for inhaled asbestos fibers, Environ Health, vol.7, issue.1, p.4, 2008.

K. Donaldson, F. A. Murphy, R. Duffin, and C. A. Poland, Asbestos, carbon nanotubes and the pleural mesothelium: a review and the hypothesis regarding the role of long fibre retention in the parietal pleura, inflammation and mesothelioma, Part Fibre Toxicol, vol.7, issue.1, p.5, 2010.

N. S. Wang, Anatomy of the pleura, Clin Chest Med, vol.19, issue.2, pp.229-269, 1998.

N. S. Wang, The preformed stomas connecting the pleural cavity and the lymphatics in the parietal pleura, Am Rev Respir Dis, vol.111, issue.1, pp.12-20, 1975.

S. P. Hammar, The pathology of benign and malignant pleural disease, Chest Surg Clin N Am, vol.4, issue.3, pp.405-435, 1994.

F. Feith, J. , and J. Mc,

, Revue de pneumologie clinique, vol.69, pp.358-62, 2013.

R. R. Mercer, J. F. Scabilloni, A. F. Hubbs, L. Wang, L. A. Battelli et al., Extrapulmonary transport of MWCNT following inhalation exposure, Part Fibre Toxicol, vol.10, issue.1, p.38, 2013.

D. W. Porter, A. F. Hubbs, R. R. Mercer, N. Wu, M. G. Wolfarth et al., Mouse pulmonary dose-and time course-responses induced by exposure to multi-walled carbon nanotubes, Toxicology, vol.269, issue.2-3, pp.136-183, 2010.

G. Oberdorster and U. Graham, Predicting EMP hazard: Lessons from studies with inhaled fibrous and non-fibrous nano-and micro-particles, Toxicol Appl Pharmacol, vol.361, pp.50-61, 2018.

S. I. Sinis, C. Hatzoglou, K. I. Gourgoulianis, and S. G. Zarogiannis, Carbon Nanotubes and Other Engineered Nanoparticles Induced Pathophysiology on Mesothelial Cells and Mesothelial Membranes. Front Physiol, vol.9, p.295, 2018.

A. A. Shvedova, N. Yanamala, E. R. Kisin, A. V. Tkach, A. R. Murray et al., Long-Term Effects of Carbon Containing Engineered Nanomaterials and Asbestos in the Lung: One Year Post Exposure Comparisons, Am J Physiol Lung Cell Mol Physiol, vol.306, issue.2, pp.170-82, 2014.

P. F. Holt, Transport of inhaled dust to extrapulmonary sites, J Pathol, vol.133, issue.2, pp.123-132, 1981.

K. M. Muller, I. Schmitz, and K. Konstantinidis, Black spots of the parietal pleura: morphology and formal pathogenesis, Respiration, vol.69, issue.3, pp.261-268, 2002.

K. Mitchev, P. Dumortier, D. Vuyst, and P. , Black Spots' and hyaline pleural plaques on the parietal pleura of 150 urban necropsy cases, Am J Surg Pathol, vol.26, issue.9, pp.1198-206, 2002.

F. D. Pooley, Proceedings: The recognition of various types of asbestos as minerals, and in tissues, Clin Sci Mol Med, vol.47, issue.3, pp.11-13, 1974.

R. F. Dodson, M. F. O'sullivan, J. Huang, D. B. Holiday, and S. P. Hammar, Asbestos in extrapulmonary sites: omentum and mesentery, Chest, vol.117, issue.2, pp.486-93, 2000.

C. Boutin, P. Dumortier, F. Rey, J. R. Viallat, and P. Devuyst, Black spots concentrate oncogenic asbestos fibers in the parietal pleura: thoracoscopic and mineralogic study, Amer J Respir Crit Care Med, vol.153, issue.1, pp.444-453, 1996.

J. C. Pairon, F. Laurent, M. Rinaldo, B. Clin, P. Andujar et al., Pleural plaques and the risk of pleural mesothelioma, J Natl Cancer Inst, vol.105, issue.4, pp.293-301, 2013.

P. Boffetta, Epidemiology of peritoneal mesothelioma: a review, Ann Oncol, vol.18, issue.6, pp.985-90, 2007.

B. Price and A. Ware, Time trend of mesothelioma incidence in the United States and projection of future cases: An update based on SEER data for 1973 through, Crit Rev Toxicol, vol.39, issue.7, pp.576-88, 2005.

K. Straif, L. Benbrahim-tallaa, R. Baan, Y. Grosse, B. Secretan et al., A review of human carcinogens--part C: metals, arsenic, dusts, and fibres, Lancet Oncol, vol.10, issue.5, pp.453-457, 2009.

, Arsenic, metals, fibres, and dusts. A review of human carcinogens 69372 Lyon Cedex 08, France: International Agency for Research on Cancer, 2012.

C. Paris, I. Thaon, F. Herin, B. Clin, A. Lacourt et al., Occupational Asbestos Exposure and Incidence of Colon and Rectal Cancers in French Men: The Asbestos-Related Diseases Cohort (ARDCo-Nut), Environ Health Perspect, vol.125, issue.3, pp.409-424, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01526878

M. Micallef, C. Shield, K. D. Vignat, J. Baldi, I. Charbotel et al., Cancers in France in 2015 attributable to occupational exposures, Int J Hyg Environ Health, vol.222, issue.1, pp.22-31, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02159287

B. Clin, I. Thaon, M. Boulanger, P. Brochard, S. Chamming's et al., Cancer of the esophagus and asbestos exposure, Am J Ind Med, vol.60, issue.11, pp.968-75, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-02092951

S. E. Mutsaers, The mesothelial cell, Int J Biochem Cell Biol, vol.36, issue.1, pp.9-16, 2004.

K. N. Michailova and K. G. Usunoff, Normal structure, development and experimental pathology, Adv Anat Embryol Cell Biol, vol.183, pp.1-144, 2006.

H. Batra and A. Vb, Pleural mesothelial cells in pleural and lung diseases, Journal of thoracic disease, vol.7, issue.6, pp.964-80, 2015.

S. E. Mutsaers, D. Whitaker, and J. M. Papadimitriou, Mesothelial regeneration is not dependent on subserosal cells, J Pathol, vol.190, pp.86-92, 2000.

S. E. Mutsaers, Mesothelial cells: their structure, function and role in serosal repair, Respirology, vol.7, issue.3, pp.171-91, 2002.

A. J. Foley-comer, S. E. Herrick, T. Al-mishlab, C. M. Prele, G. J. Laurent et al., Evidence for incorporation of free-floating mesothelial cells as a mechanism of serosal healing, J Cell Sci, vol.115, pp.1383-1392, 2002.

A. Kienzle, A. B. Servais, A. B. Ysasi, B. C. Gibney, C. D. Valenzuela et al., Free-Floating Mesothelial Cells in Pleural Fluid After Lung Surgery, Front Med (Lausanne), vol.5, p.89, 2018.

, Some nanomaterials and some fibres. 69372 Lyon Cedex 08, France: International Agency for Research on Cancer, 2017.

J. C. Wagner and G. Berry, Mesotheliomas in rats following inoculation with asbestos, British Journal of Cancer, vol.23, pp.567-81, 1969.

J. M. Davis, Structural variations between pleural and peritoneal mesotheliomas produced in rats by the injection of crocidolite asbestos, Ann Anat Pathol (Paris), vol.21, issue.2, pp.199-210, 1976.

J. M. Davis, The histopathology and ultrastructure of pleural mesotheliomas produced in the rat by injections of crocidolite asbestos, Br J Exp Pathol, vol.60, issue.6, pp.642-52, 1979.

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.3799-805, 2003.

I. Adamson and J. Bakowska, KGF and HGF are growth factors for mesothelial cells in pleural lavage fluid after intratracheal asbestos, Exp Lung Res, vol.27, pp.605-621, 2001.

T. R. Gelzleichter, E. Bermudez, J. B. Mangum, B. A. Wong, O. R. Moss et al., Pulmonary and pleural responses in Fischer 344 rats following short-term inhalation of a synthetic vitreous fiber .2. Pathobiologic responses, Fund Appl Toxicol, vol.30, issue.1, pp.39-46, 1996.

J. I. Everitt, T. R. Gelzleichter, E. Bermudez, J. B. Mangum, B. A. Wong et al., Comparison of pleural responses of rats and hamsters to subchronic inhalation of refractory ceramic fibers, Environ Health Perspect, vol.105, issue.5, pp.1209-1222, 1997.

J. Xu, D. B. Alexander, M. Futakuchi, T. Numano, K. Fukamachi et al., Size-and shapedependent pleural translocation, deposition, fibrogenesis, and mesothelial proliferation by multiwalled carbon nanotubes, Cancer Sci, vol.105, issue.7, pp.763-772, 2014.

D. Liao, Q. Wang, J. He, D. B. Alexander, M. Abdelgied et al., Persistent Pleural Lesions and Inflammation by Pulmonary Exposure of Multiwalled Carbon Nanotubes, Chem Res Toxicol, vol.31, issue.10, pp.1025-1056, 2018.

B. L. Libbus and J. E. Craighead, Chromosomal translocations with specific breakpoints in asbestos-induced rat mesotheliomas, Cancer Res, vol.48, pp.6455-61, 1988.

K. Unfried, C. Schürkes, and J. Abel, Distinct spectrum of mutations induced by crocidolite asbestos : clue for 8-hydroxydeoxyguanosine-dependent mutagenesis in vivo, Cancer Res, vol.62, pp.99-104, 2002.

C. Schurkes, W. Brock, J. Abel, and K. Unfried, Induction of 8-hydroxydeoxyguanosine by man made vitreous fibres and crocidolite asbestos administered intraperitoneally in rats, Mutat Res, vol.553, issue.1-2, pp.59-65, 2004.

R. Yamaguchi, T. Hirano, Y. Ootsuyama, S. Asami, Y. Tsurudome et al., Increased 8-hydroxyguanine in DNA and its repair activity in hamster and rat lung after intratracheal instillation of crocidolite asbestos, Jpn J Cancer Res, vol.90, issue.5, pp.505-514, 1999.

Z. Ni, Y. Q. Liu, N. Keshava, G. Zhou, W. Z. Whong et al., Analysis of K-ras and p53 mutations in mesotheliomas from humans and rats exposed to asbestos, Mutat Res, vol.468, pp.87-92, 2000.

K. Unfried, N. Kociok, M. Roller, J. Friemann, F. Pott et al., P53 mutations in tumours induced by intraperitoneal injection of crocidolite asbestos and benzo[a]pyrene in rats, Exp Toxicol Pathol, vol.49, issue.3-4, pp.181-188, 1997.

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, issue.4, pp.407-423, 2016.

M. C. Jarvis, D. Ebrahimi, N. A. Temiz, and R. S. Harris, Mutation Signatures Including APOBEC in Cancer Cell Lines, JNCI Cancer Spectr, vol.2, issue.1, 2018.

W. Liu, J. D. Ernst, and V. C. Broaddus, Phagocytosis of crocidolite asbestos induces oxidative stress, DNA damage, and apoptosis in mesothelial cells, Am J Respir cell Mol Biol, vol.23, pp.371-379, 2000.

A. Shukla, M. Gulumian, T. K. Hei, D. Kamp, Q. Rahman et al., Multiple roles of oxidants in the pathogenesis of asbestos-induced diseases, Free Radic Biol Med, vol.34, issue.9, pp.1117-1146, 2003.

L. Jiang, H. Nagai, H. Ohara, S. Hara, M. Tachibana et al., Characteristics and modifying factors of asbestos-induced oxidative DNA damage, Cancer Sci, vol.99, issue.11, pp.2142-51, 2008.

S. X. Huang, M. C. Jaurand, D. W. Kamp, J. Whysner, and T. K. Hei, Role of mutagenicity in asbestos fiber-induced carcinogenicity and other diseases, J Toxicol Environ Health B Crit Rev, vol.14, issue.1-4, pp.179-245, 2011.

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, issue.2, pp.96-118, 2013.

M. Sayan and B. T. Mossman, The NLRP3 inflammasome in pathogenic particle and fibreassociated lung inflammation and diseases, Part Fibre Toxicol, vol.13, issue.1, p.51, 2016.

Y. J. Arnoldussen, V. Skaug, M. Aleksandersen, E. Ropstad, K. H. Anmarkrud et al., Inflammation in the pleural cavity following injection of multi-walled carbon nanotubes is dependent on their characteristics and the presence of IL-1 genes, Nanotoxicology, vol.12, issue.6, pp.522-560, 2018.

N. Yanamala, E. R. Kisin, D. W. Gutkin, M. R. Shurin, M. Harper et al., Characterization of pulmonary responses in mice to asbestos/asbestiform fibers using gene expression profiles, J Toxicol Environ Health A, vol.81, issue.4, pp.60-79, 2018.

J. G. Teeguarden, B. J. Webb-robertson, K. M. Waters, A. R. Murray, E. R. Kisin et al., Comparative proteomics and pulmonary toxicity of instilled single-walled carbon nanotubes, crocidolite asbestos, and ultrafine carbon black in mice, Toxicol Sci, vol.120, issue.1, pp.123-158, 2011.

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 (Ink4a/Arf), Curr Biol, vol.27, issue.21, pp.3302-3316, 2017.

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, issue.1, p.396, 2017.

J. Port and D. J. Murphy, Mesothelioma: Identical Routes to Malignancy from Asbestos and Carbon Nanotubes, Curr Biol, vol.27, issue.21, pp.1173-1179, 2017.

F. Huaux, V. De-bousies, M. A. Parent, M. Orsi, F. Uwambayinema et al., Mesothelioma response to carbon nanotubes is associated with an early and selective accumulation of immunosuppressive monocytic cells, Part Fibre Toxicol, vol.13, issue.1, p.46, 2016.

N. Kumagai-takei, M. Maeda, Y. Chen, H. Matsuzaki, S. Lee et al., Asbestos induces reduction of tumor immunity, Clin Dev Immunol, p.481439, 2011.

M. Maeda, Y. Nishimura, N. Kumagai, H. Hayashi, T. Hatayama et al., Dysregulation of the immune system caused by silica and asbestos, J Immunotoxicol, vol.7, issue.4, pp.268-78, 2010.

D. Jean and M. C. Jaurand, Mesotheliomas in Genetically Engineered Mice Unravel Mechanism of Mesothelial Carcinogenesis, International journal of molecular sciences, vol.19, issue.8, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02478607

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, issue.24, pp.3151-60, 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, issue.3, pp.261-71, 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, issue.9, pp.2836-2880, 2016.

E. Sementino, C. W. Menges, Y. Kadariya, S. Peri, J. Xu et al., Inactivation of Tp53 and Pten drives rapid development of pleural and peritoneal malignant mesotheliomas, J Cell Physiol, 2018.

M. Nasu, M. Emi, S. Pastorino, M. Tanji, A. Powers et al., High Incidence of Somatic BAP1 alterations in sporadic malignant mesothelioma, J Thorac Oncol, vol.10, issue.4, pp.565-76, 2015.

M. Bott, M. Brevet, B. S. Taylor, S. Shimizu, T. Ito et al., The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma, Nat Genet, vol.43, issue.7, pp.668-72, 2011.

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.331-339, 2002.

D. A. Altomare, C. A. Vaslet, K. L. Skele, D. Rienzo, A. Devarajan et al., A mouse model recapitulating molecular features of human mesothelioma, Cancer Res, vol.65, issue.18, pp.8090-8095, 2005.

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, issue.12, pp.1862-1871, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01928388

D. A. Altomare, C. W. Menges, J. Pei, L. Zhang, K. L. Skele-stump et al., Activated TNF-alpha/NF-kappaB signaling via down-regulation of Fas-associated factor 1 in asbestos-induced mesotheliomas from Arf knockout mice, Proc Natl Acad Sci, vol.106, issue.9, pp.3420-3425, 2009.

J. M. Marsella, B. L. Liu, C. A. Vaslet, and A. B. Kane, Susceptibility of p53-deficient mice to induction of mesothelioma by crocidolite asbestos fibers. Environ Health paerspect, vol.105, pp.1069-72, 1997.

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 asbestosinduced mesothelioma, BMC Cancer, vol.15, p.983, 2015.

A. Kane, D. Jean, and M. C. Jaurand, Mechanism of mesothelial carcinogenesis, Occupational Cancers, 2014.

M. C. Jaurand, L. Kheuang, L. Magne, and J. Bignon, Chromosomal changes induced by chrysotile fibres or benzo(3-4)pyrene in rat pleural mesothelial cells, Mutat Res, vol.169, pp.141-149, 1986.

S. Achard, M. Perderiset, and M. C. Jaurand, Sister chromatid exchanges in rat pleural mesothelial cells treated with crocidolite, attapulgite or benzo 3-4 pyrene, Br J Ind Med, vol.44, pp.281-284, 1987.

N. S. Wang, M. C. Jaurand, L. Magne, L. Kheuang, M. C. Pinchon et al., The interactions between asbestos fibers and metaphase chromosomes of rat pleural mesothelial cells in culture. A scanning and transmission electron microscopic study, Am J Pathol, vol.126, pp.343-352, 1987.

M. Yegles, L. Saint-etienne, A. Renier, X. Janson, and M. C. Jaurand, Induction of metaphase and anaphase/telophase abnormalities by asbestos fibers in rat pleural mesothelial cells in vitro, Amer J Respir Cell Mol Biol, vol.9, issue.2, pp.186-91, 1993.

M. Yegles, X. Janson, H. Y. Dong, A. Renier, and M. C. Jaurand, Role of fibre characteristics on cytotoxicity and induction of anaphase/telophase aberrations in rat pleural mesothelial cells in vitro. Correlations with in vivo animal findings, Carcinogenesis, vol.16, issue.11, pp.2751-2759, 1995.

V. Levresse, A. Renier, J. Fleury-feith, F. Levy, S. Moritz et al., Analysis of cell cycle disruptions in cultures of rat pleural mesothelial cells exposed to asbestos fibres, Amer J Respir Cell Mol Biol, vol.17, pp.660-71, 1997.

V. Levresse, A. Renier, F. Levy, V. C. Broaddus, and M. C. Jaurand, DNA breakage in asbestos-treated normal and transformed (TSV40) rat pleural mesothelial cells, Mutagenesis, vol.15, issue.3, pp.239-283, 2000.

J. R. Pietruska and A. B. Kane, SV40 oncoproteins enhance asbestos-induced DNA double-strand breaks and abrogate senescence in murine mesothelial cells, Cancer Res, vol.67, issue.8, pp.3637-3682, 2007.

A. Renier, F. Levy, F. Pilliere, and M. C. Jaurand, Uncheduled DNA synthesis in rat pleural mesothelial cells treated with mineral fibres or benzo[a]pyrene, Mutat Res, vol.241, pp.361-368, 1990.

H. Y. Dong, A. Buard, A. Renier, F. Levy, L. Saint-etienne et al., Role of oxygen derivatives in the cytotoxicity and DNA damage produced by asbestos on rat pleural mesothelial cells in vitro, Carcinogenesis, vol.15, issue.6, pp.1251-1256, 1994.

H. Y. Dong, A. Buard, F. Levy, A. Renier, F. Laval et al., Synthesis of poly(ADP-ribose) in asbestos treated rat pleural mesothelial cells in culture, Mutat Res, vol.331, pp.197-204, 1995.

H. Fung, Y. W. Kow, B. Van-houten, and B. T. Mossman, Patterns of 8-hydroxydeoxyguanosine formation in DNA and indications of oxidative stress in rat and human pleural mesothelial cells after exposure to crocidolite asbestos, Carcinogenesis, vol.18, issue.4, pp.825-857, 1997.

C. L. Zanella, J. Posada, T. R. Tritton, and B. T. Mossman, Asbestos causes stimulation of the extracellular signal-regulated kinase 1 mitogen-activated protein kinase cascade after phosophorylation of the epidermal growth factor receptor, Cancer Res, vol.56, pp.5334-5342, 1996.

S. P. Faux, C. E. Houghton, A. Hubbard, and G. Patrick, Increased expression of epidermal growth factor receptor in rat pleural mesothelial cells correlates with carcinogenicity of mineral fibres, Carcinogenesis, vol.12, pp.2275-80, 2000.

P. B. Kopnin, I. V. Kravchenko, V. A. Furalyov, L. N. Pylev, and B. P. Kopnin, Cell type-specific effects of asbestos on intracellular ROS levels, DNA oxidation and G1 cell cycle checkpoint, Oncogene, vol.23, issue.54, pp.8834-8874, 2004.

V. C. Broaddus, Asbestos, the mesothelial cell and malignancy: a matter of life or death, Am J Respir Cell Mol Biol, vol.17, issue.6, pp.657-666, 1997.

M. M. Acencio, B. Soares, E. Marchi, C. S. Silva, L. R. Teixeira et al., Inflammatory Cytokines Contribute to Asbestos-Induced Injury of Mesothelial Cells, Lung, vol.193, issue.5, pp.831-838, 2015.

A. Bononi, C. Giorgi, S. Patergnani, D. Larson, K. Verbruggen et al., BAP1 regulates IP3R3-mediated Ca(2+) flux to mitochondria suppressing cell transformation, Nature, vol.546, issue.7659, pp.549-53, 2017.

J. K. Thompson, M. B. Macpherson, S. L. Beuschel, and A. Shukla, Asbestos-Induced Mesothelial to Fibroblastic Transition Is Modulated by the Inflammasome, Am J Pathol, vol.187, issue.3, pp.665-78, 2017.

E. Casalone, A. Allione, C. Viberti, B. Pardini, S. Guarrera et al., DNA methylation profiling of asbestos-treated MeT5A cell line reveals novel pathways implicated in asbestos response, Archives of toxicology, vol.92, issue.5, pp.1785-95, 2018.

P. Nymark, P. M. Lindholm, M. V. Korpela, L. Lahti, S. Ruosaari et al., Gene expression profiles in asbestos-exposed epithelial and mesothelial lung cell lines, BMC Genomics, vol.8, p.62, 2007.

A. Shukla, M. B. Macpherson, J. Hillegass, M. E. Ramos-nino, V. Alexeeva et al., Alterations in Gene Expression in Human Mesothelial Cells Correlate with Mineral Pathogenicity, Am J Respir Cell Mol Biol, 2009.

H. Wang, A. Gillis, C. Zhao, E. Lee, J. Wu et al., Crocidolite asbestos-induced signal pathway dysregulation in mesothelial cells, Mutat Res, vol.723, issue.2, pp.171-177, 2011.

B. Burmeister, T. Schwerdtle, I. Poser, E. Hoffmann, A. Hartwig et al., Effects of asbestos on initiation of DNA damage, induction of DNA-strand breaks, P53-expression and apoptosis in primary, SV40-transformed and malignant human mesothelial cells, Mutat Res, vol.558, issue.1-2, pp.81-92, 2004.

W. Lohcharoenkal, L. Wang, T. A. Stueckle, C. Z. Dinu, V. Castranova et al., Chronic exposure to carbon nanotubes induces invasion of human mesothelial cells through matrix metalloproteinase-2, ACS Nano, vol.7, issue.9, pp.7711-7734, 2013.

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, p.59, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01073918

P. G. Coin, V. L. Roggli, and A. R. Brody, Persistence of long, thin chrysotile asbestos fibers in the lungs of rats, Environ Health Perspect, vol.102, issue.5, pp.197-206, 1994.

L. Greillier and P. Astoul, Mesothelioma and asbestos-related pleural diseases, Respiration, vol.76, issue.1, pp.1-15, 2008.

L. Fazzo, G. Minelli, D. Santis, M. Bruno, C. Zona et al., Epidemiological surveillance of mesothelioma mortality in Italy, Cancer epidemiology, vol.55, pp.184-91, 2018.

J. A. Merchant, Human epidemiology: a review of fiber type and characteristics in the development of malignant and nonmalignant disease, Environ Health Perspect, vol.88, pp.287-93, 1990.

Y. I. Baris and P. Grandjean, Prospective study of mesothelioma mortality in Turkish villages with exposure to fibrous zeolite, J Natl Cancer Inst, vol.98, issue.6, pp.414-421, 2006.

, Asbestos: Selected Cancers, IOM, 2006.

B. Fubini, Surface reactivity in the pathogenic response to particulates, Environ health Perspect, vol.105, issue.5, pp.1013-1033, 1997.

J. C. Mcdonald, Epidemiology of malignant mesothelioma--an outline, Ann Occup Hyg, vol.54, issue.8, pp.851-858, 2010.

J. C. Mcdonald, J. Harris, and B. Armstrong, Mortality in a cohort of vermiculite miners exposed to fibrous amphibole in Libby, Montana, Occup Environ Med, vol.61, issue.4, pp.363-369, 2004.

, Silica and some silicates, IARC Monogr Eval Carcinog Risk Chem Hum, vol.42, pp.1-239, 1987.

V. M. Harik, Geometry of carbon nanotubes and mechanisms of phagocytosis and toxic effects, Toxicol Lett, vol.273, pp.69-85, 2017.

M. C. Jaurand, Use of in-vitro genotoxicity and cell transformation assays to evaluate potential carcinogenicity of fibres, Mechanisms in Fiber Carcinogenesis. 1401996, pp.55-72

T. Hei, D. Louie, and Y. L. Zhao, Genotoxicity versus carcinogenicity: Implications from fiber toxicity studies, Inhal Toxicol, vol.12, issue.s3, pp.141-148, 2000.

R. A. Maccorkle, S. D. Slattery, D. R. Nash, and B. R. Brinkley, Intracellular protein binding to asbestos induces aneuploidy in human lung fibroblasts, Cell Motil Cytoskeleton, vol.63, issue.10, pp.646-57, 2006.

E. R. Kisin, A. R. Murray, L. Sargent, D. Lowry, M. Chirila et al., Genotoxicity of carbon nanofibers: are they potentially more or less dangerous than carbon nanotubes or asbestos?, Toxicol Appl Pharmacol, vol.252, issue.1, pp.1-10, 2011.

J. Muller, I. Decordier, P. H. Hoet, N. Lombaert, L. Thomassen et al., Clastogenic and aneugenic effects of multi-wall carbon nanotubes in epithelial cells, Carcinogenesis, vol.29, issue.2, pp.427-460, 2008.

H. K. Lindberg, G. C. Falck, S. Suhonen, M. Vippola, E. Vanhala et al., Genotoxicity of nanomaterials: DNA damage and micronuclei induced by carbon nanotubes and graphite nanofibres in human bronchial epithelial cells in vitro, Toxicol Lett, vol.186, pp.166-73, 2009.

F. Cammisuli, S. Giordani, A. Gianoncelli, C. Rizzardi, L. Radillo et al., Iron-related toxicity of single-walled carbon nanotubes and crocidolite fibres in human mesothelial cells investigated by Synchrotron XRF microscopy, Scientific reports, vol.8, issue.1, p.706, 2018.

G. Liu, R. Beri, A. Mueller, and D. W. Kamp, Molecular mechanisms of asbestos-induced lung epithelial cell apoptosis, Chem Biol Interact

B. Fubini and L. Mollo, Role of iron in the reactivity of mineral fibers, Toxicol Lett, vol.82, issue.3, pp.951-60, 1995.

D. Van-berlo, M. J. Clift, C. Albrecht, and R. P. Schins, Carbon nanotubes: an insight into the mechanisms of their potential genotoxicity, Swiss Med Wkly, vol.142, p.13698, 2012.

D. Bernstein, V. Castranova, K. Donaldson, B. Fubini, J. Hadley et al., Testing of fibrous particles: short-term assays and strategies. of an ILSI Risk Science Institute Working Group Inhal Toxicol, vol.17, pp.497-537, 2005.

D. W. Berman and K. S. Crump, A meta-analysis of asbestos-related cancer risk that addresses fiber size and mineral type, Crit Rev Toxicol, vol.38, pp.49-73, 2008.

M. Casali, M. Carugno, A. Cattaneo, D. Consonni, C. Mensi et al., Asbestos Lung Burden in Necroscopic Samples from the General Population of, Ann Occup Hyg, vol.59, issue.7, pp.909-930, 2015.

E. Merler, A. Somigliana, P. Girardi, and P. G. Barbieri, Residual fibre lung burden among patients with pleural mesothelioma who have been occupationally exposed to asbestos, Occup Environ Med, vol.74, issue.3, pp.218-245, 2017.

S. Pollastri, A. F. Gualtieri, R. Vigliaturo, K. Ignatyev, E. Strafella et al., Stability of mineral fibres in contact with human cell cultures. An in situ muXANES, muXRD and XRF iron mapping study, Chemosphere, vol.164, pp.547-57, 2016.

Y. Song, J. Thiagarajah, and A. S. Verkman, Sodium and chloride concentrations, pH, and depth of airway surface liquid in distal airways, J Gen Physiol, vol.122, issue.5, pp.511-520, 2003.

Y. Suzuki, S. R. Yuen, and A. R. Short, thin asbestos fibers contribute to the development of human malignant mesothelioma: pathological evidence, Int J Hyg Environ Health, vol.208, issue.3, pp.201-211, 2005.

R. F. Dodson and S. P. Hammar, Pleural mesothelioma in a woman whose documented past exposure to asbestos was from smoking asbestos-containing filtered cigarettes: the comparative value of analytical transmission electron microscopic analysis of lung and lymph-node tissue, Inhal Toxicol, vol.18, issue.9, pp.679-84, 2006.

H. Nagai and S. Toyokuni, Biopersistent fiber-induced inflammation and carcinogenesis: lessons learned from asbestos toward safety of fibrous nanomaterials, Arch Biochem Biophys, vol.502, issue.1, pp.1-7, 2010.

N. Shinohara, T. Nakazato, K. Ohkawa, M. Tamura, N. Kobayashi et al., Longterm retention of pristine multi-walled carbon nanotubes in rat lungs after intratracheal instillation, J Appl Toxicol, vol.36, issue.4, pp.501-510, 2016.

J. F. Lechner, T. Tokiwa, M. Laveck, W. F. Benedict, S. Banks-schlegel et al., Asbestos-associated chromosomal changes in human mesothelial cells, Proc Natl Acad Sci, vol.82, issue.11, pp.3884-3892, 1985.

T. W. Hesterberg, G. A. Hart, J. Chevalier, W. C. Miiller, R. D. Hamilton et al., The importance of fiber biopersistence and lung dose in determining the chronic inhalation effects of X607, RCF1, and chrysotile asbestos in rats, Toxicol Appl Pharmacol, vol.153, pp.68-82, 1998.

Y. Kodama, C. J. Boreiko, S. C. Maness, and T. W. Hesterberg, Cytotoxic and cytogenetic effects of asbestos on human bronchial epithelial cells in culture, Carcinogenesis, vol.14, issue.4, pp.691-698, 1993.

D. W. Kamp, V. A. Israbian, A. V. Yeldandi, R. J. Panos, P. Graceffa et al., Phytic acid, an iron chelator, attenuates pulmonary inflammation and fibrosis in rats after intratracheal instillation of asbestos, Toxicol Pathol, vol.23, issue.6, pp.689-95, 1995.

A. Shukla, M. Jung, M. Stern, N. K. Fukagawa, D. J. Taatjes et al., Asbestos induces mitochondrial DNA damage and dysfunction linked to the development of apoptosis, Am J Physiol Lung Cell Mol Physiol, vol.285, issue.5, pp.1018-1043, 2003.

R. K. Srivastava, M. Lohani, A. B. Pant, and Q. Rahman, Cyto-genotoxicity of amphibole asbestos fibers in cultured human lung epithelial cell line: role of surface iron, Toxicol Ind Health, vol.26, issue.9, pp.575-82, 2010.

T. K. Hei, Z. Y. He, and K. Suzuki, Effects of antioxidants on fiber mutagenesis, Carcinogenesis, vol.16, issue.7, pp.1573-1581, 1995.

D. W. Kamp and S. A. Weitzman, The molecular basis of asbestos induced lung injury, Thorax, vol.54, issue.7, pp.638-52, 1999.

V. Valinluck and L. C. Sowers, Inflammation-mediated cytosine damage: a mechanistic link between inflammation and the epigenetic alterations in human cancers, Cancer Res, vol.67, issue.12, pp.5583-5589, 2007.

H. Kasai and K. Kawai, DNA methylation at the C-5 position of cytosine by methyl radicals: a possible role for epigenetic change during carcinogenesis by environmental agents, Chem Res Toxicol, vol.22, issue.6, pp.984-993, 2009.

S. Reuter, S. C. Gupta, M. M. Chaturvedi, and B. B. Aggarwal, Oxidative stress, inflammation, and cancer: how are they linked?, Free Radic Biol Med, vol.49, issue.11, pp.1603-1619, 2010.

N. H. Heintz, Y. M. Janssen-heininger, and B. T. Mossman, Asbestos, lung cancers, and mesotheliomas: from molecular approaches to targeting tumor survival pathways, Am J Respir Cell Mol Biol, vol.42, issue.2, pp.133-142, 2010.

E. Kettunen, S. Knuutila, and V. K. Sarhadi, Malignant Mesothelioma: Molecular Markers, Occupational Cancers

Y. Yoshikawa, M. Emi, T. Hashimoto-tamaoki, M. Ohmuraya, A. Sato et al., Highdensity array-CGH with targeted NGS unmask multiple noncontiguous minute deletions on chromosome 3p21 in mesothelioma, Proc Natl Acad Sci, vol.113, issue.47, pp.13432-13439, 2016.

Y. Sekido, Genomic abnormalities and signal transduction dysregulation in malignant mesothelioma cells, Cancer Sci, vol.101, issue.1, pp.1-6, 2010.

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, issue.5, pp.1323-1357, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02478599

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, issue.12, pp.1548-65, 2018.

P. Andujar, J. Wang, A. Descatha, F. Galateau-sallé, A. Abd-alsamad et al., p16 INK4A inactivation mechanisms in non small-cell lung cancer patients occupationally exposed to asbestos, Lung Cancer, vol.67, issue.1, pp.23-30, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00388198

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, issue.8, pp.1555-1564, 2008.

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, issue.1, pp.227-261, 2009.

S. Toyooka, K. O. Toyooka, R. Maruyama, A. K. Virmani, L. Girard et al., DNA methylation profiles of lung tumors, Mol Cancer Ther, vol.1, issue.1, pp.61-68, 2001.

T. Hirao, R. Bueno, C. J. Chen, G. J. Gordon, E. Heilig et al., Alterations of the p16INK4 locus in human malignant mesothelial tumors, Carcinogenesis, vol.23, pp.1127-1157, 2002.

L. Wong, J. Zhou, D. Anderson, and R. A. Kratzke, Inactivation of p16INK4a expression in malignant mesothelioma by methylation, Lung Cancer, vol.38, issue.2, pp.131-137, 2002.

C. J. Marsit, E. A. Houseman, B. C. Christensen, K. Eddy, R. Bueno et al., Examination of a CpG island methylator phenotype and implications of methylation profiles in solid tumors, Cancer Res, vol.66, issue.21, pp.10621-10630, 2006.

A. Destro, G. L. Ceresoli, E. Baryshnikova, I. Garassino, P. A. Zucali et al., Gene methylation in pleural mesothelioma: correlations with clinico-pathological features and patient's follow-up, Lung Cancer, vol.59, issue.3, pp.369-76, 2008.

M. Guled, L. Lahti, P. M. Lindholm, K. Salmenkivi, I. Bagwan et al., CDKN2A, NF2, and JUN are dysregulated among other genes by miRNAs in malignant mesothelioma -A miRNA microarray analysis, Genes Chromosomes Cancer, vol.48, issue.7, pp.615-638, 2009.

P. Andujar, C. Lecomte, A. Renier, J. Fleury-feith, L. Kheuang et al., Clinicopathological features and somatic gene alterations in refractory ceramic fibre-induced murine mesothelioma reveal mineral fibre-induced mesothelioma identities, Carcinogenesis, vol.28, issue.7, pp.1599-605, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00329999

O. Mor, P. Yaron, M. Huszar, A. Yellin, O. Jakobovitz et al., Absence of p53 mutations in malignant mesothelioma, Am J Respir Cell Mol Biol, vol.16, pp.9-13, 1997.

F. Kitamura, S. Araki, T. Tanigawa, H. Miura, H. Akabane et al., Assessment of mutations of Ha-and Ki-ras oncogenes and the p53 suppressor gene in seven malignant mesothelioma patients exposed to asbestos. PCR-SSCP and sequencing analyses of paraffin-embedded primary tumors Ind Health, vol.36, pp.52-58, 1998.

P. Andujar, J. C. Pairon, A. Renier, A. Descatha, I. Hysi et al., Differential mutation profiles and similar intronic TP53 polymorphisms in asbestos-related lung cancer and pleural mesothelioma, Mutagenesis, vol.28, issue.3, pp.323-354, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02478590

A. B. Bianchi, S. Mitsunaga, J. Cheng, W. Klein, S. C. Jhanwar et al., High frequency of inactivating mutations in the neurofibromatosis type 2 gene (NF2) in primary malignant mesothelioma, Proc Natl Acad Sci, USA, vol.92, pp.10854-10862, 1995.

Y. Sekido, H. I. Pass, S. Bader, D. J. Mew, M. F. Christmas et al., Neurofibromatosis type 2 (NF2) gene is somatically mutated in mesothelioma but not in lung cancer, Cancer Res, vol.55, pp.1227-1258, 1995.

A. Miyanaga, M. Masuda, K. Tsuta, K. Kawasaki, Y. Nakamura et al., Hippo pathway gene mutations in malignant mesothelioma: revealed by RNA and targeted exon sequencing, J Thorac Oncol, vol.10, issue.5, pp.844-51, 2015.

Y. Sekido, Molecular pathogenesis of malignant mesothelioma, Carcinogenesis, vol.34, issue.7, pp.1413-1422, 2013.

I. Stamenkovic and Q. Yu, Merlin, a "magic" linker between extracellular cues and intracellular signaling pathways that regulate cell motility, proliferation, and survival, Curr Protein Pept Sci, vol.11, issue.6, pp.471-84, 2010.

S. S. Murthy and J. R. Testa, Asbestos, chromosomal deletions, and tumor suppressor gene alterations in human malignant mesothelioma, J Cell Physiol, vol.180, pp.150-157, 1999.

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, issue.2, pp.881-94, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00531343

D. Lallemand, M. Curto, I. Saotome, M. Giovannini, and A. I. Mcclatchey, NF2 deficiency promotes tumorigenesis and metastasis by destabilizing adherens junctions, Genes Dev, vol.17, issue.9, pp.1090-100, 2003.

C. Yi, S. Troutman, D. Fera, A. Stemmer-rachamimov, J. L. Avila et al., A Tight Junction-Associated Merlin-Angiomotin Complex Mediates Merlin's Regulation of Mitogenic Signaling and Tumor Suppressive Functions, Cancer Cell, vol.19, issue.4, pp.527-567, 2010.

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, issue.10, pp.1022-1027, 2011.

S. Sneddon, J. S. Leon, I. M. Dick, G. Cadby, N. Olsen et al., Absence of germline mutations in BAP1 in sporadic cases of malignant mesothelioma, Gene, vol.563, issue.1, pp.103-108, 2015.

V. Panou, M. Vyberg, U. M. Weinreich, C. Meristoudis, U. G. Falkmer et al., The established and future biomarkers of malignant pleural mesothelioma, Cancer Treat Rev, vol.41, issue.6, pp.486-95, 2015.

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, issue.28, pp.3748-452, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02478602

M. C. Jaurand and D. Jean, Biomolecular Pathways and Malignant Pleural Mesothelioma, Malignant Pleural Mesothelioma: Present Status and Future Directions, pp.173-96, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02479956

M. Hylebos, G. Van-camp, J. P. Van-meerbeeck, O. De-beeck, and K. , The Genetic Landscape of Malignant Pleural Mesothelioma: Results from Massively Parallel Sequencing, J Thorac Oncol, vol.11, issue.10, pp.1615-1641, 2016.

X. L. Liu, R. Zuo, and W. B. Ou, The hippo pathway provides novel insights into lung cancer and mesothelioma treatment, J Cancer Res Clin Oncol, vol.144, issue.11, pp.2097-106, 2018.

T. Sato and Y. Sekido, NF2/Merlin Inactivation and Potential Therapeutic Targets in Mesothelioma, International journal of molecular sciences, vol.19, issue.4, p.988, 2018.

E. Felley-bosco and R. Stahel, Hippo/YAP pathway for targeted therapy, Translational lung cancer research, vol.3, issue.2, pp.75-83, 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, issue.12, pp.3191-202, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-02478604

H. Murakami, T. Mizuno, T. Taniguchi, M. Fujii, F. Ishiguro et al., LATS2 Is a Tumor Suppressor Gene of Malignant Mesothelioma, Cancer Res, vol.71, issue.3, pp.873-83, 2011.

C. Thurneysen, I. Opitz, S. Kurtz, W. Weder, R. A. Stahel et al., Functional inactivation of NF2/merlin in human mesothelioma, Lung Cancer, vol.64, issue.2, pp.140-147, 2009.

S. Romagnoli, E. Fasoli, V. Vaira, M. Falleni, C. Pellegrini et al., Identification of potential therapeutic targets in malignant mesothelioma using cell-cycle gene expression analysis, Am J Pathol, vol.174, issue.3, pp.762-70, 2009.

S. Crispi, C. Fagliarone, A. Biroccio, D. Angelo, C. Galati et al., Antiproliferative effect of Aurora kinase targeting in mesothelioma, Lung Cancer, vol.70, issue.3, pp.271-280, 2010.

O. D. Roe, E. Anderssen, H. Sandeck, T. Christensen, E. Larsson et al., Malignant pleural mesothelioma: genome-wide expression patterns reflecting general resistance mechanisms and a proposal of novel targets, Lung Cancer, vol.67, issue.1, pp.57-68, 2010.

F. Lopez-rios, S. Chuai, R. Flores, S. Shimizu, T. Ohno et al., Global gene expression profiling of pleural mesotheliomas: overexpression of aurora kinases and P16/CDKN2A deletion as prognostic factors and critical evaluation of microarray-based prognostic prediction, Cancer Res, vol.66, issue.6, pp.2970-2979, 2006.

C. I. Rubin and G. F. Atweh, The role of stathmin in the regulation of the cell cycle, J Cell Biochem, vol.93, issue.2, pp.242-50, 2004.

J. Y. Kim, C. Harvard, L. You, Z. Xu, K. Kuchenbecker et al., Stathmin is overexpressed in malignant mesothelioma, Anticancer Res, vol.27, issue.1A, pp.39-44, 2007.

K. A. Birnie, Y. Y. Yip, D. C. Ng, M. B. Kirschner, G. Reid et al., Loss of mir-223 and JNK Signalling Contribute to Elevated Stathmin in Malignant Pleural Mesothelioma, Mol Cancer Res, vol.13, issue.7, pp.1106-1124, 2015.

B. J. Aubrey, A. Strasser, and G. L. Kelly, Tumor-Suppressor Functions of the TP53 Pathway, Cold Spring Harb Perspect Med, vol.6, issue.5, 2016.

M. Carbone, H. Yang, H. I. Pass, T. Krausz, J. R. Testa et al., BAP1 and cancer, Nat Rev Cancer, vol.13, issue.3, pp.153-162, 2013.

C. Luchini, L. D. Wood, L. Cheng, A. Nottegar, B. Stubbs et al., Extranodal extension of lymph node metastasis is a marker of poor prognosis in oesophageal cancer: a systematic review with meta-analysis, J Clin Pathol, 2016.

T. A. Knijnenburg, L. Wang, M. T. Zimmermann, N. Chambwe, G. F. Gao et al., Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas, Cell Rep, vol.23, issue.1, pp.239-54, 2018.

F. D. Mairinger, R. Werner, E. Flom, J. Schmeller, S. Borchert et al., miRNA regulation is important for DNA damage repair and recognition in malignant pleural mesothelioma, Virchows Arch, vol.470, issue.6, pp.627-664, 2017.

Y. Soini, V. Kinnula, R. Kaarteenaho-wiik, E. Kurttila, K. Linnainmaa et al., Apoptosis and expression of apoptosis regulating proteins bcl-2, mcl-1, bcl-X, and bax in malignant mesothelioma, Clin Cancer Res, vol.5, issue.11, pp.3508-3523, 1999.

S. L. O'kane, R. J. Pound, A. Campbell, N. Chaudhuri, M. J. Lind et al., Expression of bcl-2 family members in malignant pleural mesothelioma, Acta Oncol, vol.45, issue.4, pp.449-53, 2006.

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 and Differentiation, vol.16, issue.8, pp.1146-55, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00388174

L. Jin, V. J. Amatya, Y. Takeshima, L. Shrestha, K. Kushitani et al., Evaluation of apoptosis and immunohistochemical expression of the apoptosis-related proteins in mesothelioma, Hiroshima J Med Sci, vol.59, issue.2, pp.27-33, 2010.

L. E. Leard and V. C. Broaddus, Mesothelial cell proliferation and apoptosis, Respirology, vol.9, issue.3, pp.292-301, 2004.

S. M. Wilson, D. Barbone, T. M. Yang, D. M. Jablons, R. Bueno et al., mTOR mediates survival signals in malignant mesothelioma grown as tumor fragment spheroids, Am J Respir Cell Mol Biol, vol.39, issue.5, pp.576-83, 2008.

G. Kafiri, D. M. Thomas, N. A. Shepherd, T. Krausz, D. P. Lane et al., p53 expression is common in malignant mesothelioma, Histopathology, vol.21, issue.4, pp.331-335, 1992.

M. Ramael, G. Lemmens, C. Eerdekens, C. Buysse, I. Deblier et al., Immunoreactivity for p53 protein in malignant mesothelioma and non-neoplastic mesothelium, J Pathol, vol.168, pp.371-376, 1992.

F. G. Mayall, H. Goddard, and A. R. Gibbs, The Frequency of p53 Immunostaining in Asbestos-Associated Mesotheliomas and Non-Asbestos-Associated Mesotheliomas

, Histopathology, vol.22, issue.4, pp.383-389, 1993.

R. L. Attanoos, A. Griffin, and A. R. Gibbs, The use of immunohistochemistry in distinguishing reactive from neoplastic mesothelium. A novel use for desmin and comparative evaluation with epithelial membrane antigen, p53, platelet-derived growth factorreceptor, P-glycoprotein and Bcl-2, Histopathology, vol.43, issue.3, pp.231-239, 2003.

Z. Feng and A. J. Levine, The regulation of energy metabolism and the IGF-1/mTOR pathways by the p53 protein, Trends Cell Biol, vol.20, issue.7, pp.427-461, 2010.

S. Singhal, R. Wiewrodt, L. D. Malden, K. M. Amin, K. Matzie et al., Gene expression profiling of malignant mesothelioma, Clin Cancer Res, vol.9, issue.8, pp.3080-97, 2003.

Y. Y. Lei, W. J. Wang, J. H. Mei, and C. L. Wang, Mitogen-activated protein kinase signal transduction in solid tumors, Asian Pac J Cancer Prev, vol.15, issue.20, pp.8539-8587, 2014.

Y. Ohta, V. Shridhar, R. K. Bright, G. P. Kalemkerian, W. Du et al., VEGF and VEGF type C play an important role in angiogenesis and lymphangiogenesis in human malignant mesothelioma tumours, Br J Cancer, vol.81, issue.1, pp.54-61, 1999.

J. Konig, E. Tolnay, T. Wiethege, and K. Muller, Co-expression of vascular endothelial growth factor and its receptor flt-1 in malignant pleural mesothelioma, Respiration, vol.67, pp.36-40, 2000.

L. Strizzi, A. Catalano, G. Vianale, S. Orecchia, A. Casalini et al., Vascular endothelial growth factor is an autocrine growth factor in human malignant mesothelioma, J Pathol, vol.193, pp.468-75, 2001.

A. L. Filho, F. Baltazar, C. Bedrossian, C. Michael, and F. C. Schmitt, Immunohistochemical expression and distribution of VEGFR-3 in malignant mesothelioma, Diagn Cytopathol, vol.35, issue.12, pp.786-91, 2007.

A. Y. Lee, D. J. Raz, B. He, and D. M. Jablons, Update on the molecular biology of malignant mesothelioma, Cancer, vol.109, issue.8, pp.1454-61, 2007.

R. Masood, A. Kundra, S. Zhu, G. Xia, P. Scalia et al., Malignant mesothelioma growth inhibition by agents that target the VEGF and VEGF-C autocrine loops, Int J Cancer, vol.104, issue.5, pp.603-613, 2003.

A. Jacobson, J. Brinck, M. J. Briskin, A. P. Spicer, and P. Heldin, Expression of human hyaluronan synthases in response to external stimuli, Biochem J, vol.348, pp.29-35, 2000.

P. Heldin, T. Asplund, D. Ytterberg, S. Thelin, and T. C. Laurent, Characterization of the molecular mechanism involved in the activation of hyaluronan synthetase by platelet-derived growth factor in human mesothelial cells, Biochem J, vol.283, pp.165-70, 1992.

B. I. Gerwin, J. F. Lechner, R. R. Reddel, A. B. Roberts, K. C. Robbins et al., Comparison of production of transforming growth factor-beta and platelet-derived growth factor by normal human mesothelial cells and mesothelioma cell lines, Cancer Res, vol.47, issue.23, pp.6180-6184, 1987.

M. A. Versnel, L. Claessonwelsh, A. Hammacher, M. J. Bouts, T. H. Vanderkwast et al., Human malignant mesothelioma cell lines express PDGF beta-receptors whereas cultured normal mesothelial cells express predominantly PDGF alpha-receptors, Oncogene, vol.6, issue.11, pp.2005-2016, 1991.

L. J. Metheny-barlow, F. B. Van-gijssel, H. E. Marrogi, A. Gerwin, and B. I. , Paradoxical effects of platelet-derived growth factor-A overexpression in malignant mesothelioma. Antiproliferative effects in vitro and tumorigenic stimulation in vivo, Am J Respir Cell Mol Biol, vol.24, issue.6, pp.694-702, 2001.

A. Van-der-meeren, M. B. Seddon, C. A. Betsholtz, J. F. Lechner, and B. I. Gerwin, Tumorigenic conversion of human mesothelial cells as a consequence of platelet-derived growth factor-A chain overexpression, Amer J Respir Cell Mol Biol, vol.8, issue.2, pp.214-235, 1993.

M. Honda, T. Kanno, Y. Fujita, A. Gotoh, T. Nakano et al., Mesothelioma cell proliferation through autocrine activation of PDGF-betabeta receptor, Cell Physiol Biochem, vol.29, issue.5-6, pp.667-74, 2012.

V. Agarwal, M. J. Lind, and L. Cawkwell, Targeted epidermal growth factor receptor therapy in malignant pleural mesothelioma: Where do we stand?, Cancer Treat Rev, vol.37, issue.7, pp.533-575, 2011.

C. D. Hoang, X. Zhang, P. D. Scott, T. J. Guillaume, M. A. Maddaus et al., Selective activation of insulin receptor substrate-1 and -2 in pleural mesothelioma cells: association with distinct malignant phenotypes, Cancer Res, vol.64, issue.20, pp.7479-85, 2004.

B. A. Whitson and R. A. Kratzke, Molecular pathways in malignant pleural mesothelioma, Cancer Lett, vol.239, issue.2, pp.183-192, 2006.

M. C. Jaurand, J. Fleury-feith, and . Cells, Textbook of pleural diseases, pp.27-37, 2008.

T. C. Lee, Y. Zhang, C. Aston, R. Hintz, J. Jagirdar et al., Normal human mesothelial cells and mesothelioma cell lines express insulin-like growth factor I and associated molecules, Cancer Res, vol.53, issue.12, pp.2858-64, 1993.

Z. Liu and J. Klominek, Regulation of matrix metalloprotease activity in malignant mesothelioma cell lines by growth factors, Thorax, vol.58, issue.3, pp.198-203, 2003.

T. Thayaparan, J. F. Spicer, and J. Maher, The role of the HGF/Met axis in mesothelioma, Biochemical Society transactions, vol.44, issue.2, pp.363-70, 2016.

P. Harvey, A. Warn, S. Dobbin, N. Arakaki, Y. Daikuhara et al., Expression of HGF/SF in mesothelioma cell lines and its effects on cell motility, proliferation and morphology, Br J Cancer, vol.77, pp.1052-1061, 1998.

T. Mukohara, G. Civiello, I. J. Davis, M. L. Taffaro, J. Christensen et al., Inhibition of the met receptor in mesothelioma, Clin Cancer Res, vol.11, issue.22, pp.8122-8152, 2005.

R. Jagadeeswaran, P. C. Ma, T. Y. Seiwert, S. Jagadeeswaran, O. Zumba et al., Functional analysis of c-Met/hepatocyte growth factor pathway in malignant pleural mesothelioma, Cancer Res, vol.66, issue.1, pp.352-61, 2006.

K. Kawaguchi, H. Murakami, T. Taniguchi, M. Fujii, S. Kawata et al., Combined inhibition of MET and EGFR suppresses proliferation of malignant mesothelioma cells, Carcinogenesis, vol.30, issue.7, pp.1097-105, 2009.

L. Vintman, S. Nielsen, A. Berner, R. Reich, and B. Davidson, Mitogen-activated protein kinase expression and activation does not differentiate benign from malignant mesothelial cells, Cancer, vol.103, issue.11, pp.2427-2460, 2005.

M. De-melo, M. W. Gerbase, J. Curran, and J. C. Pache, Phosphorylated extracellular signalregulated kinases are significantly increased in malignant mesothelioma, J Histochem Cytochem, vol.54, issue.8, pp.855-61, 2006.

R. Eguchi, Y. Fujimori, H. Takeda, C. Tabata, T. Ohta et al., Arsenic trioxide induces apoptosis through JNK and ERK in human mesothelioma cells, J Cell Physiol, vol.226, issue.3, pp.762-770, 2011.

W. B. Ou, C. Hubert, J. M. Corson, R. Bueno, D. L. Flynn et al., Targeted inhibition of multiple receptor tyrosine kinases in mesothelioma, Neoplasia, vol.13, issue.1, pp.12-22, 2011.

S. Zhou, L. Liu, H. Li, G. Eilers, Y. Kuang et al., Multipoint targeting of the PI3K/mTOR pathway in mesothelioma, Br J Cancer, vol.110, issue.10, pp.2479-88, 2014.

A. Besson, S. M. Robbins, and V. W. Yong, PTEN/MMAC1/TEP1 in signal transduction and tumorigenesis, Eur J Biochem, vol.263, issue.3, pp.605-616, 1999.

D. A. Altomare, H. You, G. H. Xiao, M. E. Ramos-nino, K. L. Skele et al., Human and mouse mesotheliomas exhibit elevated AKT/PKB activity, which can be targeted pharmacologically to inhibit tumor cell growth, Oncogene, vol.24, issue.40, pp.6080-6089, 2005.

Y. Suzuki, H. Murakami, K. Kawaguchi, T. Taniguchi, M. Fujii et al., Activation of the PI3K-AKT pathway in human malignant mesothelioma cells, Molecular Medicine Reports, vol.2, issue.2, pp.181-189, 2009.

M. R. Makena, A. Ranjan, V. Thirumala, and A. P. Reddy, Cancer stem cells: Road to therapeutic resistance and strategies to overcome resistance, Biochim Biophys Acta Mol Basis Dis, 2018.

N. Takebe, L. Miele, P. J. Harris, W. Jeong, H. Bando et al., Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update, Nat Rev Clin Oncol, vol.12, issue.8, pp.445-64, 2015.

H. Clevers, Wnt/beta-catenin signaling in development and disease, Cell, vol.127, issue.3, pp.469-80, 2006.

A. Y. Lee, B. He, L. You, Z. Xu, J. Mazieres et al., Dickkopf-1 antagonizes Wnt signaling independent of beta-catenin in human mesothelioma, Biochem Biophys Res Commun, vol.323, issue.4, pp.1246-50, 2004.

B. He, A. Y. Lee, S. Dadfarmay, L. You, Z. Xu et al., Secreted frizzled-related protein 4 is silenced by hypermethylation and induces apoptosis in beta-catenin-deficient human mesothelioma cells, Cancer Res, vol.65, issue.3, pp.743-751, 2005.

S. Batra, Y. Shi, K. M. Kuchenbecker, B. He, N. Reguart et al., Wnt inhibitory factor-1, a Wnt antagonist, is silenced by promoter hypermethylation in malignant pleural mesothelioma, Biochem Biophys Res Commun, vol.342, issue.4, pp.1228-1260, 2006.

H. Kohno, V. J. Amatya, Y. Takeshima, K. Kushitani, N. Hattori et al., Aberrant promoter methylation of WIF-1 and SFRP1, 2, 4 genes in mesothelioma, Oncol Rep, vol.24, issue.2, pp.423-454, 2010.

J. Mazieres, L. You, B. He, Z. Xu, S. Twogood et al., Wnt2 as a new therapeutic target in malignant pleural mesothelioma, Int J Cancer, vol.117, issue.2, pp.326-358, 2005.

E. Felley-bosco, I. Opitz, and M. Meerang, Hedgehog Signaling in Malignant Pleural Mesothelioma, Genes, vol.6, issue.3, pp.500-511, 2015.

H. A. Kim, M. C. Kim, N. Y. Kim, and Y. Kim, Inhibition of hedgehog signaling reduces the side population in human malignant mesothelioma cell lines. Cancer gene therapy, vol.22, pp.387-95, 2015.

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, issue.9, pp.1269-83, 2018.

C. B. Lim, C. M. Prele, H. M. Cheah, Y. Y. Cheng, S. Klebe et al., Mutational analysis of hedgehog signaling pathway genes in human malignant mesothelioma, PLoS ONE, vol.8, issue.6, p.66685, 2013.

M. Rossini, P. Rizzo, I. Bononi, A. Clementz, R. Ferrari et al., New Perspectives on Diagnosis and Therapy of Malignant Pleural Mesothelioma, Frontiers in oncology, vol.8, p.91, 2018.

M. A. Dawson and T. Kouzarides, Cancer epigenetics: from mechanism to therapy, Cell, vol.150, issue.1, pp.12-27, 2012.

K. C. Mcloughlin, A. S. Kaufman, and D. S. Schrump, Targeting the epigenome in malignant pleural mesothelioma, Translational lung cancer research, vol.6, issue.3, pp.350-65, 2017.

F. Vandermeers, N. Sriramareddy, S. Costa, C. Hubaux, R. Cosse et al., The role of epigenetics in malignant pleural mesothelioma, Lung Cancer, vol.81, issue.3, pp.311-319, 2013.

D. M. Pardoll, The blockade of immune checkpoints in cancer immunotherapy, Nat Rev Cancer, vol.12, issue.4, pp.252-64, 2012.

L. A. Lievense, D. H. Sterman, R. Cornelissen, and J. G. Aerts, Checkpoint Blockade in Lung Cancer and Mesothelioma, Am J Respir Crit Care Med, vol.196, issue.3, pp.274-82, 2017.

J. Minnema-luiting, H. Vroman, J. Aerts, and R. Cornelissen, Heterogeneity in Immune Cell Content in Malignant Pleural Mesothelioma, International journal of molecular sciences, vol.19, issue.4, 2018.

A. Guazzelli, E. Bakker, M. Krstic-demonacos, M. P. Lisanti, F. Sotgia et al., Anti-CTLA-4 therapy for malignant mesothelioma, Immunotherapy, vol.9, issue.3, pp.273-80, 2017.

K. Oehl, B. Vrugt, I. Opitz, and M. Meerang, Heterogeneity in Malignant Pleural Mesothelioma, International journal of molecular sciences, vol.19, issue.6, 2018.

F. Galateau-salle, A. Churg, V. Roggli, and W. D. Travis, The 2015 World Health Organization Classification of Tumors of the Pleura: Advances since the 2004 Classification, J Thorac Oncol, vol.11, issue.2, pp.142-54, 2016.

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, issue.1, pp.89-108, 2018.

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.

G. J. Gordon, G. N. Rockwell, R. V. Jensen, J. G. Rheinwald, J. N. Glickman et al., Identification of novel candidate oncogenes and tumor suppressors in malignant pleural mesothelioma using large-scale transcriptional profiling, Am J Pathol, vol.166, issue.6, pp.1827-1867, 2005.

E. Felley-bosco, Special Issue on Mechanisms of Mesothelioma Heterogeneity: Highlights and Open Questions, International journal of molecular sciences, vol.19, issue.11, 2018.

D. Hanahan and R. A. Weinberg, The hallmarks of cancer, Cell, vol.100, pp.57-70, 2000.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, issue.5, pp.646-74, 2012.