C. Arrowsmith, C. Bountra, P. Fish, K. Lee, and M. Schapira, Epigenetic protein families: a new frontier for drug discovery, Nature Reviews Drug Discovery, vol.13, issue.5, pp.384-400, 2012.
DOI : 10.1038/nrd3674

M. Dawson and T. Kouzarides, Cancer Epigenetics: From Mechanism to Therapy, Cell, vol.150, issue.1, pp.12-27, 2012.
DOI : 10.1016/j.cell.2012.06.013

M. Suva, N. Riggi, and B. Bernstein, Epigenetic Reprogramming in Cancer, Science, vol.339, issue.6127, pp.1567-70, 2013.
DOI : 10.1126/science.1230184

V. Dewoskin and R. Million, The epigenetics pipeline, Nature Reviews Drug Discovery, vol.12, issue.9, pp.661-663, 2013.
DOI : 10.1038/nrd4091

D. Feng, J. Wu, Y. Tian, H. Zhou, Y. Zhou et al., Targeting of Histone Deacetylases to Reactivate Tumour Suppressor Genes and Its Therapeutic Potential in a Human Cervical Cancer Xenograft Model, PLoS ONE, vol.137, issue.11, p.80657, 2013.
DOI : 10.1371/journal.pone.0080657.t003

T. Wagner, P. Brand, T. Heinzel, and O. Krämer, Histone deacetylase 2 controls p53 and is a critical factor in tumorigenesis, Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, vol.1846, issue.2, pp.524-562, 2014.
DOI : 10.1016/j.bbcan.2014.07.010

Y. Ma, J. H. Wang, X. Shin, V. Chen, D. Chen et al., Histone deacetylase 3 inhibits new tumor suppressor gene DTWD1 in gastric cancer, Am J Cancer Res, vol.5, pp.663-73, 2015.

M. Duvic and J. Vu, Vorinostat: a new oral histone deacetylase inhibitor approved for cutaneous T-cell lymphoma, Expert Opinion on Investigational Drugs, vol.19, issue.7, pp.1111-1131, 2007.
DOI : 10.1038/nrd2133

C. Grant, F. Rahman, R. Piekarz, C. Peer, R. Frye et al., Romidepsin: a new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors, Expert Review of Anticancer Therapy, vol.10, issue.7, pp.997-1008, 2010.
DOI : 10.1586/era.10.88

R. Poole, Belinostat: First Global Approval, Drugs, vol.103, issue.1, pp.1543-54, 2014.
DOI : 10.1007/s40265-014-0275-8

M. Fenichel, FDA Approves New Agent for Multiple Myeloma, JNCI Journal of the National Cancer Institute, vol.107, issue.6, p.165, 2015.
DOI : 10.1093/jnci/djv165

J. How, M. Minden, L. Brian, E. Chen, J. Brandwein et al., A phase I trial of two sequence-specific schedules of decitabine and vorinostat in patients with acute myeloid leukemia, Leukemia & Lymphoma, vol.110, issue.1, pp.2793-802, 2015.
DOI : 10.1111/bjh.13016

R. Robey, A. Chakraborty, A. Basseville, V. Luchenko, J. Bahr et al., Histone Deacetylase Inhibitors: Emerging Mechanisms of Resistance, Molecular Pharmaceutics, vol.8, issue.6, pp.2021-2052, 2011.
DOI : 10.1021/mp200329f

S. Mithraprabhu, T. Khong, and A. Spencer, Overcoming inherent resistance to histone deacetylase inhibitors in multiple myeloma cells by targeting pathways integral to the actin cytoskeleton, Cell Death and Disease, vol.83, issue.3, p.1134, 2014.
DOI : 10.1007/s00018-011-0892-2

N. Azad, C. Zahnow, C. Rudin, and S. Baylin, The future of epigenetic therapy in solid tumours???lessons from the past, Nature Reviews Clinical Oncology, vol.7, issue.5, pp.256-66, 2013.
DOI : 10.1038/nrclinonc.2013.42

R. Momparler, S. Cote, L. Momparler, and Y. Idaghdour, Epigenetic therapy of acute myeloid leukemia using 5-aza-2'-deoxycytidine (decitabine) in combination with inhibitors of histone methylation and deacetylation, Clinical Epigenetics, vol.6, issue.1, p.19, 2014.
DOI : 10.1186/1471-2407-13-60

M. Kalac, L. Scotto, E. Marchi, J. Amengual, V. Seshan et al., HDAC inhibitors and decitabine are highly synergistic and associated with unique gene-expression and epigenetic profiles in models of DLBCL, Blood, vol.118, issue.20, pp.5506-5522, 2011.
DOI : 10.1182/blood-2011-02-336891

T. Ahrens, S. Timme, J. Hoeppner, J. Ostendorp, S. Hembach et al., Selective inhibition of esophageal cancer cells by combination of HDAC inhibitors and Azacytidine, Epigenetics, vol.46, issue.5, pp.431-476, 2015.
DOI : 10.1093/nar/gks1055

T. Sun, Y. Zhang, B. Pang, D. Hyun, M. Yang et al., Engineered Nanoparticles for Drug Delivery in Cancer Therapy, Angewandte Chemie International Edition, vol.6, pp.12320-64, 2014.
DOI : 10.1002/anie.201403036

F. Bahhaj, F. Dekker, N. Martinet, and P. Bertrand, Delivery of epidrugs, Drug Discovery Today, vol.19, issue.9, pp.1337-52, 2014.
DOI : 10.1016/j.drudis.2014.03.017

URL : https://hal.archives-ouvertes.fr/hal-01201998

Y. Wang, J. Byrne, M. Napier, and J. Desimone, Engineering nanomedicines using stimuli-responsive biomaterials, Advanced Drug Delivery Reviews, vol.64, issue.11, pp.1021-1051, 2012.
DOI : 10.1016/j.addr.2012.01.003

Q. Yin, J. Shen, Z. Zhang, H. Yu, and Y. Li, Reversal of multidrug resistance by stimuli-responsive drug delivery systems for therapy of tumor, Advanced Drug Delivery Reviews, vol.65, issue.13-14, pp.1699-715, 2013.
DOI : 10.1016/j.addr.2013.04.011

H. Maeda, Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity, Advanced Drug Delivery Reviews, vol.91, pp.3-6, 2015.
DOI : 10.1016/j.addr.2015.01.002

K. Miki, K. Oride, S. Inoue, Y. Kuramochi, R. Nayak et al., Ring-opening metathesis polymerization-based synthesis of polymeric nanoparticles for enhanced tumor imaging in vivo: Synergistic effect of folate-receptor targeting and PEGylation, Biomaterials, vol.31, issue.5, pp.934-976, 2010.
DOI : 10.1016/j.biomaterials.2009.10.005

J. Johnson, Y. Lu, A. Burts, Y. Lim, M. Finn et al., -Azide Bivalent-Bottle-Brush Polymers by ROMP: Grafting-Through and Clicking-To, Journal of the American Chemical Society, vol.133, issue.3, pp.559-66, 2011.
DOI : 10.1021/ja108441d

URL : https://hal.archives-ouvertes.fr/jpa-00210025

F. Gueugnon, I. Denis, D. Pouliquen, F. Collette, R. Delatouche et al., Nanoparticles Produced by Ring-Opening Metathesis Polymerization Using Norbornenyl-poly(ethylene oxide) as a Ligand-Free Generic Platform for Highly Selective In Vivo Tumor Targeting, Biomacromolecules, vol.14, issue.7, pp.2396-402, 2013.
DOI : 10.1021/bm400516b

URL : https://hal.archives-ouvertes.fr/hal-00916100

R. Delatouche, I. Denis, M. Grinda, F. Bahhaj, E. Baucher et al., Design of pH responsive clickable prodrugs applied to histone deacetylase inhibitors: A new strategy for anticancer therapy, European Journal of Pharmaceutics and Biopharmaceutics, vol.85, issue.3, pp.862-72, 2013.
DOI : 10.1016/j.ejpb.2013.03.006

URL : https://hal.archives-ouvertes.fr/hal-01006764

G. Scita, D. Fiore, and P. , The endocytic matrix, Nature, vol.177, issue.7280, pp.464-73, 2010.
DOI : 10.1038/nature08910

O. Arrieta, L. Medina, E. Estrada-lobato, N. Hernandez-pedro, G. Villanueva-rodriguez et al., First-line chemotherapy with liposomal doxorubicin plus cisplatin for patients with advanced malignant pleural mesothelioma: phase II trial, British Journal of Cancer, vol.13, issue.6, pp.1027-1059, 2012.
DOI : 10.1016/j.athoracsur.2009.01.026

F. Vandermeers, S. Sriramareddy, C. Costa, R. Hubaux, J. Cosse et al., The role of epigenetics in malignant pleural mesothelioma, Lung Cancer, vol.81, issue.3, pp.311-319, 2013.
DOI : 10.1016/j.lungcan.2013.05.014

L. Krug, H. Kindler, H. Calvert, C. Manegold, A. Tsao et al., Vorinostat in patients with advanced malignant pleural mesothelioma who have progressed on previous chemotherapy (VANTAGE-014): a phase 3, double-blind, randomised, placebo-controlled trial, The Lancet Oncology, vol.16, issue.4, pp.447-56, 2015.
DOI : 10.1016/S1470-2045(15)70056-2

H. Zong, D. Shah, K. Selwa, R. Tsuchida, R. Rattan et al., Design and Evaluation of Tumor-Specific Dendrimer Epigenetic Therapeutics, ChemistryOpen, vol.184, issue.3, pp.335-376, 2015.
DOI : 10.1002/open.201402141

F. Collette, R. Delatouche, C. Blanquart, F. Gueugnon, M. Grégoire et al., Easy and effective method to produce functionalized particles for cellular uptake, Journal of Polymer Science Part A: Polymer Chemistry, vol.27, issue.1, pp.176-89, 2013.
DOI : 10.1002/pola.26357

URL : https://hal.archives-ouvertes.fr/hal-00918361

C. Charrier, J. Clarhaut, J. Gesson, G. Estiu, O. Wiest et al., Synthesis and Modeling of New Benzofuranone Histone Deacetylase Inhibitors that Stimulate Tumor Suppressor Gene Expression, Journal of Medicinal Chemistry, vol.52, issue.9, pp.3112-3117, 2009.
DOI : 10.1021/jm9002439

URL : https://hal.archives-ouvertes.fr/hal-00438795

. Vorinostat, Polymer Conjugate Nanoparticles for Acid-Responsive Delivery and Passive Tumor Targeting, Biomacromolecules, vol.15, pp.4534-4577, 2014.

A. Olaharski, J. Z. Woo, J. Lim, S. Hubbard, A. Zhang et al., The Histone Deacetylase Inhibitor Trichostatin A Has Genotoxic Effects in Human Lymphoblasts In Vitro, Toxicological Sciences, vol.93, issue.2, pp.341-348, 2006.
DOI : 10.1093/toxsci/kfl068

M. Yoshida, Y. Hoshikawa, K. Koseki, K. Mori, and T. Beppu, Structural specificity for biological activity of trichostatin a, a specific inhibitor of mammalian cell cycle with potent differentiation-inducing activity in friend leukemia cells., The Journal of Antibiotics, vol.43, issue.9, pp.1101-1107, 1990.
DOI : 10.7164/antibiotics.43.1101

F. Gueugnon, P. Cartron, C. Charrier, P. Bertrand, J. Fonteneau et al., New histone deacetylase inhibitors improve cisplatin antitumor properties against thoracic cancer cells, Oncotarget, vol.5, issue.12, pp.4504-4519, 2014.
DOI : 10.18632/oncotarget.2056

P. Wang, Y. Wang, H. Hu, C. Spencer, X. Liang et al., Sequential Removal of Photolabile Protecting Groups for Carbonyls with Controlled Wavelength, The Journal of Organic Chemistry, vol.73, issue.16, pp.6152-6159, 2008.
DOI : 10.1021/jo8008275

C. Blanquart, M. François, C. Charrier, P. Bertrand, and M. Grégoire, Pharmacological Characterization of Histone Deacetylase Inhibitor and Tumor Cell-Growth Inhibition Properties of New Benzofuranone Compounds, Current Cancer Drug Targets, vol.11, issue.8, pp.919-947, 2011.
DOI : 10.2174/156800911797264761

A. Kung, Practices and Pitfalls of Mouse Cancer Models in Drug Discovery, Adv Cancer Res, vol.96, pp.191-212, 2007.
DOI : 10.1016/S0065-230X(06)96007-2

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.1105-1121, 2011.
DOI : 10.1183/09031936.00081310

S. Shaker, M. Bernstein, L. Momparler, and R. Momparler, Preclinical evaluation of antineoplastic activity of inhibitors of DNA methylation (5-aza-2???-deoxycytidine) and histone deacetylation (trichostatin A, depsipeptide) in combination against myeloid leukemic cells, Leukemia Research, vol.27, issue.5, pp.437-481, 2003.
DOI : 10.1016/S0145-2126(02)00222-9