S. Klebe, N. Brownlee, A. Mahar, J. Burchette, T. Sporn et al., Sarcomatoid mesothelioma: a clinical???pathologic correlation of 326 cases, Modern Pathology, vol.22, issue.3, pp.470-479, 2010.
DOI : 10.1046/j.1365-2559.41.s2.41.x

N. Vogelzang, J. Rusthoven, J. Symanowski, C. Denham, E. Kaukel et al., Phase III Study of Pemetrexed in Combination With Cisplatin Versus Cisplatin Alone in Patients With Malignant Pleural Mesothelioma, Journal of Clinical Oncology, vol.21, issue.14, pp.2636-2644, 2003.
DOI : 10.1200/JCO.2003.11.136

A. Rahim, S. Ho, G. Coward, and J. , The role of interleukin-6 in malignant mesothelioma, Transl Lung Cancer Res, vol.4, pp.55-66, 2015.

G. Pinton, A. Manente, D. Tavian, L. Moro, and L. Mutti, Therapies currently in Phase II trials for malignant pleural mesothelioma, Expert Opinion on Investigational Drugs, vol.57, issue.3, pp.1255-1263, 2013.
DOI : 10.1158/0008-5472.CAN-08-4523

S. Moore, L. Darlison, and A. Tod, Living with mesothelioma. A literature review, European Journal of Cancer Care, vol.7, issue.4, pp.458-468, 2010.
DOI : 10.1111/j.1365-2354.2009.01162.x

M. Deraco, D. Baratti, I. Hutanu, R. Bertuli, and S. Kusamura, The Role of Perioperative Systemic Chemotherapy in Diffuse Malignant Peritoneal Mesothelioma Patients Treated with Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy, Annals of Surgical Oncology, vol.46, issue.4, pp.1093-1100, 2013.
DOI : 10.1245/s10434-012-2845-x

S. Bölükbas, M. Eberlein, and J. Schirren, Prospective Study on Functional Results After Lung-Sparing Radical Pleurectomy in the Management of Malignant Pleural Mesothelioma, Journal of Thoracic Oncology, vol.7, issue.5, pp.900-905, 2012.
DOI : 10.1097/JTO.0b013e31824de2dc

M. New, H. Olzscha, L. Thangue, and N. , HDAC inhibitor-based therapies: Can we interpret the code?, Molecular Oncology, vol.26, issue.6, pp.637-656, 2012.
DOI : 10.1016/j.molonc.2012.09.003

F. Guillot, B. Boutin, C. Blanquart, J. Fonteneau, M. Robard et al., Vaccination with epigenetically treated mesothelioma cells induces immunisation and blocks tumour growth, Vaccine, vol.29, issue.33, pp.5534-5543, 2011.
DOI : 10.1016/j.vaccine.2011.05.029

S. Shishodia, M. Chaturvedi, and B. Aggarwal, Role of Curcumin in Cancer Therapy, Current Problems in Cancer, vol.31, issue.4, pp.243-305, 2007.
DOI : 10.1016/j.currproblcancer.2007.04.001

S. Gupta, S. Patchva, and B. Aggarwal, Therapeutic Roles of Curcumin: Lessons Learned from Clinical Trials, The AAPS Journal, vol.15, issue.1, pp.195-218, 2013.
DOI : 10.1208/s12248-012-9432-8

S. Fu and R. Kurzrock, Development of curcumin as an epigenetic agent, Cancer, vol.29, issue.pt 11, pp.4670-4676, 2010.
DOI : 10.1002/cncr.25414

G. Franci, M. Miceli, and L. Altucci, Targeting epigenetic networks with polypharmacology: a new avenue to tackle cancer, Epigenomics, vol.2, issue.6, pp.731-742, 2010.
DOI : 10.2217/epi.10.62

T. Esatbeyoglu, P. Huebbe, I. Ernst, D. Chin, A. Wagner et al., Curcumin-From Molecule to Biological Function, Angewandte Chemie International Edition, vol.7, issue.481, pp.5308-5332, 2012.
DOI : 10.1002/anie.201107724

M. Cotto, F. Cabanillas, M. Tirado, M. Garcia, and E. Pacheco, Epigenetic therapy of lymphoma using histone deacetylase inhibitors, Clinical and Translational Oncology, vol.123, issue.1, pp.401-409, 2010.
DOI : 10.1007/s12094-010-0527-3

M. Teiten, M. Dicato, and M. Diederich, Curcumin as a regulator of epigenetic events, Molecular Nutrition & Food Research, vol.126, issue.9, pp.1619-1629, 2013.
DOI : 10.1002/mnfr.201300201

F. Vahid, H. Zand, E. Nosrat-mirshekarlou, R. Najafi, and A. Hekmatdoost, The role dietary of bioactive compounds on the regulation of histone acetylases and deacetylases: A review, Gene, vol.562, issue.1, pp.8-15, 2015.
DOI : 10.1016/j.gene.2015.02.045

B. Adams, E. Ferstl, M. Davis, M. Herold, S. Kurtkaya et al., Synthesis and biological evaluation of novel curcumin analogs as anti-cancer and anti-angiogenesis agents, Bioorganic & Medicinal Chemistry, vol.12, issue.14, pp.3871-3883, 2004.
DOI : 10.1016/j.bmc.2004.05.006

D. Pouliquen, Curcumin. Synthesis, emerging role in pain management and health implications, pp.978-979
URL : https://hal.archives-ouvertes.fr/inserm-01480303

B. Aggarwal, D. Danda, S. Gupta, and P. Gehlot, Models for prevention and treatment of cancer: Problems vs promises, Biochemical Pharmacology, vol.78, issue.9, pp.1083-1094, 2009.
DOI : 10.1016/j.bcp.2009.05.027

P. Workman, E. Aboagye, F. Balkwill, A. Balmain, G. Bruder et al., Guidelines for the welfare and use of animals in cancer research, British Journal of Cancer, vol.63, issue.11, pp.1555-1577, 2010.
DOI : 10.1101/gad.1771409

E. De-bree, H. Rosing, J. Michalakis, J. Romanos, K. Relakis et al., Intraperitoneal chemotherapy with taxanes for ovarian cancer with peritoneal dissemination, European Journal of Surgical Oncology (EJSO), vol.32, issue.6, pp.666-670, 2006.
DOI : 10.1016/j.ejso.2006.03.008

A. Khar, A. Ali, B. Pardhasaradhi, Z. Begum, and R. Anjum, Antitumor activity of curcumin is mediated through the induction of apoptosis in AK-5 tumor cells, FEBS Letters, vol.1317, issue.1, pp.165-168, 1999.
DOI : 10.1016/S0014-5793(99)00114-3

A. Zanotto-filho, K. Coradini, E. Braganhol, R. Schröder, C. Oliveira et al., Curcumin-loaded lipid-core nanocapsules as a strategy to improve pharmacological efficacy of curcumin in glioma treatment, European Journal of Pharmaceutics and Biopharmaceutics, vol.83, issue.2, pp.156-67, 2013.
DOI : 10.1016/j.ejpb.2012.10.019

Y. Wang, A. Rishi, W. Wu, L. Polin, S. Sharma et al., Curcumin suppresses growth of mesothelioma cells in vitro and in vivo, in part, by stimulating apoptosis, Molecular and Cellular Biochemistry, vol.269, issue.1-2, pp.83-94, 2011.
DOI : 10.1007/s11010-011-0878-2

Y. Yamauchi, Y. Izumi, K. Asakura, Y. Hayashi, and H. Nomori, Curcumin Induces Autophagy in ACC-MESO-1 Cells, Phytotherapy Research, vol.119, issue.1-2, pp.1779-1783, 2012.
DOI : 10.1002/ptr.4645

J. Miller, J. Thompson, M. Macpherson, S. Beuschel, C. Westbom et al., Curcumin: A Double Hit on Malignant Mesothelioma, Cancer Prevention Research, vol.7, issue.3, pp.330-340, 2014.
DOI : 10.1158/1940-6207.CAPR-13-0259

L. Mayol, C. Serri, C. Menale, S. Crispi, M. Piccolo et al., Curcumin loaded PLGA???poloxamer blend nanoparticles induce cell cycle arrest in mesothelioma cells, European Journal of Pharmaceutics and Biopharmaceutics, vol.93, pp.37-45, 2015.
DOI : 10.1016/j.ejpb.2015.03.005

D. Pouliquen, B. Nawrocki-raby, A. Ouacher, C. Deleine, S. Blandin et al., New prospects with curcumin in the treatment of mesothelioma: lessons from an orthotopic rat tumor model. In : Curcumin. Synthesis, emerging role in pain management and health implications, pp.978-979

D. Roulois, S. Deshayes, M. Guilly, J. 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.34664-34687, 2016.
DOI : 10.18632/oncotarget.8970

URL : https://hal.archives-ouvertes.fr/inserm-01306838

E. Levy-nissenbaum, W. Khan, R. Pawar, R. Tabakman, E. Naftali et al., Pharmacokinetic and efficacy study of cisplatin and paclitaxel formulated in a new injectable poly(sebacic-co-ricinoleic acid) polymer, European Journal of Pharmaceutics and Biopharmaceutics, vol.82, issue.1, pp.85-93, 2012.
DOI : 10.1016/j.ejpb.2012.06.004

B. Aggarwal, S. Banerjeee, U. Bharadwaj, B. Sung, S. Shishodia et al., RETRACTED: Curcumin induces the degradation of cyclin E expression through ubiquitin-dependent pathway and up-regulates cyclin-dependent kinase inhibitors p21 and p27 in multiple human tumor cell lines, Biochemical Pharmacology, vol.73, issue.7, pp.1024-1032, 2007.
DOI : 10.1016/j.bcp.2006.12.010

J. Chen, F. Wang, and W. Chen, Modulation of apoptosis-related cell signalling pathways by curcumin as a strategy to inhibit tumor progression, Molecular Biology Reports, vol.28, issue.10, pp.4583-4594, 2014.
DOI : 10.1007/s11033-014-3329-9

M. Shanmugam, G. Rane, M. Kanchi, F. Arfuso, A. Chinnathambi et al., The Multifaceted Role of Curcumin in Cancer Prevention and Treatment, Molecules, vol.20, issue.2, pp.2728-2769, 2015.
DOI : 10.3390/molecules20022728

S. Gupta, S. Prasad, J. Kim, S. Patchva, L. Webb et al., Multitargeting by curcumin as revealed by molecular interaction studies, Natural Product Reports, vol.579, issue.Suppl. 2, pp.1937-1955, 2011.
DOI : 10.1039/c1np00051a

C. Lu, E. Song, D. Hu, M. Chen, C. Xue et al., Curcumin Induces Cell Death in Human Uveal Melanoma Cells through Mitochondrial Pathway, Current Eye Research, vol.595, issue.4, pp.352-360, 2010.
DOI : 10.1016/j.freeradbiomed.2008.12.006

M. Romero-hernández, P. Eguía-aguilar, M. Perézpeña-diazconti, A. Rodríguez-leviz, S. Sadowinski-pine et al., Toxic effects induced by curcumin in human astrocytoma cell lines, Toxicology Mechanisms and Methods, vol.18, issue.9, pp.650-659, 2013.
DOI : 10.3109/15376516.2013.826768

C. Clark, M. Mceachern, S. Shah, Y. Rong, X. Rong et al., Curcumin Inhibits Carcinogen and Nicotine-Induced Mammalian Target of Rapamycin Pathway Activation in Head and Neck Squamous Cell Carcinoma, Cancer Prevention Research, vol.3, issue.12, pp.1586-1595, 2010.
DOI : 10.1158/1940-6207.CAPR-09-0244

L. Li, F. Braiteh, and R. Kurzrock, Liposome-encapsulated curcumin, Cancer, vol.890, issue.6, pp.1322-1331, 2005.
DOI : 10.1002/cncr.21300

P. Killian, E. Kronski, K. Michalik, O. Barbieri, S. Astigiano et al., Curcumin inhibits prostate cancer metastasis in vivo by targeting the inflammatory cytokines CXCL1 and -2, Carcinogenesis, vol.33, issue.12, pp.2507-2519, 2012.
DOI : 10.1093/carcin/bgs312

A. Kunwar, A. Barik, B. Mishra, K. Rathinasamy, R. Pandey et al., Quantitative cellular uptake, localization and cytotoxicity of curcumin in normal and tumor cells, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1780, issue.4, pp.673-679, 2008.
DOI : 10.1016/j.bbagen.2007.11.016

L. Aparicio, M. Blanco, R. Castosa, C. Á. Valladares, M. Calvo et al., Clinical implications of epithelial cell plasticity in cancer progression, Cancer Letters, vol.366, issue.1, pp.1-10, 2015.
DOI : 10.1016/j.canlet.2015.06.007

Y. Lin, Y. Liu, N. Tsai, J. Hsieh, C. Chen et al., A Lipo-PEG-PEI complex for encapsulating curcumin that enhances its antitumor effects on curcumin-sensitive and curcumin-resistance cells, Nanomedicine: Nanotechnology, Biology and Medicine, vol.8, issue.3, pp.318-327, 2012.
DOI : 10.1016/j.nano.2011.06.011

E. Turrini, L. Ferruzzi, and C. Fimognari, Natural compounds to overcome cancer chemoresistance: toxicological and clinical issues, Expert Opinion on Drug Metabolism & Toxicology, vol.2012, issue.1, pp.1677-1690, 2014.
DOI : 10.1007/s00204-011-0675-4

Y. Li and T. Zhang, Targeting cancer stem cells by curcumin and clinical applications, Cancer Letters, vol.346, issue.2, pp.197-205, 2014.
DOI : 10.1016/j.canlet.2014.01.012

R. Mirmezami, B. Moran, K. Harvey, T. Cecil, K. Chandrakumaran et al., Cytoreductive surgery and intraperitoneal chemotherapy for colorectal peritoneal metastases, World Journal of Gastroenterology, vol.20, issue.38, pp.14018-14032, 2014.
DOI : 10.3748/wjg.v20.i38.14018

D. Wang, M. Veena, K. Stevenson, C. Tang, B. Ho et al., Liposome-Encapsulated Curcumin Suppresses Growth of Head and Neck Squamous Cell Carcinoma In vitro and in Xenografts through the Inhibition of Nuclear Factor ??B by an AKT-Independent Pathway, Clinical Cancer Research, vol.14, issue.19, pp.6228-6236, 2008.
DOI : 10.1158/1078-0432.CCR-07-5177

W. Orr, J. Denbo, K. Saab, A. Myers, C. Ng et al., RETRACTED: Liposome-encapsulated curcumin suppresses neuroblastoma growth through nuclear factor-kappa B inhibition, Surgery, vol.151, issue.5, pp.736-744, 2012.
DOI : 10.1016/j.surg.2011.12.014

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3331939

S. Alam, J. Panda, and V. Chauhan, Novel dipeptide nanoparticles for effective curcumin delivery, Int J Nanomedicine, vol.7, pp.4207-4222, 2012.

N. David, The Pharmacology of Dimethyl Sulfoxide, Annual Review of Pharmacology, vol.12, issue.1, pp.353-374, 1972.
DOI : 10.1146/annurev.pa.12.040172.002033

R. Lind, C. Begay, and A. Gandolfi, Hepatoprotection by Dimethyl Sulfoxide, Toxicology and Applied Pharmacology, vol.166, issue.2, pp.145-150, 2000.
DOI : 10.1006/taap.2000.8949

A. Issa, S. Volate, and M. Wargovich, The role of phytochemicals in inhibition of cancer and inflammation: New directions and perspectives, Journal of Food Composition and Analysis, vol.19, issue.5, pp.405-419, 2006.
DOI : 10.1016/j.jfca.2006.02.009

D. Pouliquen, C. Olivier, E. Hervouet, F. Pedelaborde, E. Debien et al., Dietary prevention of malignant glioma aggressiveness, implications in oxidant stress and apoptosis, International Journal of Cancer, vol.14, issue.2, pp.288-295, 2008.
DOI : 10.1002/ijc.23513

D. Pouliquen, Hepatic mitochondrial function and brain tumours, Current Opinion in Clinical Nutrition and Metabolic Care, vol.10, issue.4, pp.475-479, 2007.
DOI : 10.1097/MCO.0b013e328108f452

M. Benvenuto, R. Mattera, G. Taffera, M. Giganti, P. Lido et al., The Potential Protective Effects of Polyphenols in Asbestos-Mediated Inflammation and Carcinogenesis of Mesothelium, Nutrients, vol.8, issue.5, 2016.
DOI : 10.3390/nu8050275

G. Jagetia and B. Aggarwal, ???Spicing Up??? of the Immune System by Curcumin, Journal of Clinical Immunology, vol.279, issue.49, pp.19-35, 2007.
DOI : 10.1007/s10875-006-9066-7

V. Ch, A. Ali, B. Pardhasaradhi, R. Srivastava, S. Singh et al., Immunomodulatory effects of curcumin: in-vivo, Int Immunopharmacol, vol.8, pp.688-700, 2008.

R. Sreejayan and . Mn, Nitric Oxide Scavenging by Curcuminoids, Journal of Pharmacy and Pharmacology, vol.146, issue.1, pp.105-107, 1997.
DOI : 10.1111/j.2042-7158.1997.tb06761.x

I. Brouet and H. Ohshima, Curcumin, an Anti-tumor Promoter and Anti-inflammatory Agent, Inhibits Induction of Nitric Oxide Synthase in Activated Macrophages, Biochemical and Biophysical Research Communications, vol.206, issue.2, pp.533-540, 1995.
DOI : 10.1006/bbrc.1995.1076

M. Chan, H. Huang, M. Fenton, and D. Fong, In Vivo Inhibition of Nitric Oxide Synthase Gene Expression by Curcumin, a Cancer Preventive Natural Product with Anti-Inflammatory Properties, Biochemical Pharmacology, vol.55, issue.12, pp.1955-1962, 1998.
DOI : 10.1016/S0006-2952(98)00114-2

S. Antony, R. Kuttan, and G. Kuttan, Immunomodulatory Activity of Curcumin, Immunological Investigations, vol.90, issue.5-6, pp.291-303, 1999.
DOI : 10.3109/08820139909062263

N. Vishvakarma, A. Kumar, A. Kumar, S. Kant, A. Bharti et al., Myelopotentiating effect of curcumin in tumor-bearing host: Role of bone marrow resident macrophages, Toxicology and Applied Pharmacology, vol.263, issue.1, pp.111-121, 2012.
DOI : 10.1016/j.taap.2012.06.004

S. Leclercq, F. Gueugnon, B. Boutin, F. Guillot, C. Blanquart et al., A 5-aza-2'-deoxycytidine/valproate combination induces cytotoxic T-cell response against mesothelioma, European Respiratory Journal, vol.38, issue.5, pp.1-12, 2011.
DOI : 10.1183/09031936.00081310

H. Yang, P. Lan, Z. Hou, Y. Guan, J. Zhang et al., Histone deacetylase inhibitor SAHA epigenetically regulates miR-17-92 cluster and MCM7 to upregulate MICA expression in hepatoma, British Journal of Cancer, vol.63, issue.1, pp.112-121, 2015.
DOI : 10.1371/journal.pone.0045133

Y. Chang, H. Chuang, C. Hsu, S. Gambhir, and J. Hwang, Immunomodulation of Curcumin on Adoptive Therapy with T Cell Functional Imaging in Mice, Cancer Prevention Research, vol.5, issue.3, pp.444-452, 2011.
DOI : 10.1158/1940-6207.CAPR-11-0308

F. Luo, X. Song, Y. Zhang, and Y. Chu, Low-dose curcumin leads to the inhibition of tumor growth via enhancing CTL-mediated antitumor immunity, International Immunopharmacology, vol.11, issue.9, pp.1234-1240, 2011.
DOI : 10.1016/j.intimp.2011.04.002

S. Bhattacharyya, D. Mandal, B. Saha, G. Sen, T. Das et al., Curcumin Prevents Tumor-induced T Cell Apoptosis through Stat-5a-mediated Bcl-2 Induction, Journal of Biological Chemistry, vol.282, issue.22, pp.15954-15964, 2007.
DOI : 10.1074/jbc.M608189200

S. Bhattacharyya, M. Sakib-hossain, D. Mohanty, S. , S. Sen et al., Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts, Cellular and Molecular Immunology, vol.174, issue.4, pp.306-315, 2010.
DOI : 10.1016/S0968-0896(02)00461-3

Z. Zhang, Y. Z. Zhang, and Z. , Distribution and characterization of tumor-associated macrophages/microglia in rat C6 glioma, Oncology Letters, vol.10, pp.2442-2446, 2015.
DOI : 10.3892/ol.2015.3533

K. So, K. Min, J. Hong, and J. Lee, Interleukin-6 expression by interactions between gynecologic cancer cells and human mesenchymal stem cells promotes epithelial-mesenchymal transition, International Journal of Oncology, vol.47, pp.1451-1459, 2015.
DOI : 10.3892/ijo.2015.3122