I. Kohno, M. Pouyssegur, and J. , Targeting the ERK signaling pathway in cancer therapy, Annals of Medicine, vol.14, issue.3
DOI : 10.1016/j.gde.2003.12.002

URL : https://hal.archives-ouvertes.fr/hal-00321058

P. J. Roberts and C. J. Der, Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer, Oncogene, vol.24, issue.22, pp.3291-3310, 2007.
DOI : 10.1038/72799

A. Tsimberidou, Targeted therapy in cancer, Cancer Chemotherapy and Pharmacology, vol.20, issue.18, pp.1113-1132, 2015.
DOI : 10.1158/1078-0432.CCR-14-0603

B. Keam, In vitro anticancer activity of PI3K alpha selective inhibitor BYL719 in head and neck cancer, Anticancer Res, vol.35, pp.175-182, 2015.

E. H. Estey, Epigenetics in clinical practice: the examples of azacitidine and decitabine in myelodysplasia and acute myeloid leukemia, Leukemia, vol.dii:pii, issue.9, pp.1803-1812, 2013.
DOI : 10.1038/leu.2011.234

N. Boissel, Th??rapeutiques cibl??es dans??les??leuc??mies aigu??s, R??animation, vol.15, issue.4, pp.278-284, 2006.
DOI : 10.1016/j.reaurg.2006.06.002

L. Degos, S. Castaigne, P. Fenaux, M. Daniel, and C. Chomienne, Le traitement des leuc??mies aigu??s ?? promy??locytes par l'acide tout-trans r??tino??que., m??decine/sciences, vol.7, issue.5, p.460, 1991.
DOI : 10.4267/10608/4386

J. M. Redman, E. M. Hill, D. Aldeghaither, and L. M. Weiner, Mechanisms of action of therapeutic antibodies for cancer, Molecular Immunology, vol.67, issue.2, pp.28-45, 2015.
DOI : 10.1016/j.molimm.2015.04.002

A. Bosly, Anticorps monoclonal anti-CD20 (rituximab) dans??les??maladies h??matologiques et??les??affections auto-immunes, R??animation, vol.15, issue.4, pp.270-277, 2006.
DOI : 10.1016/j.reaurg.2006.06.015

L. Hui and Y. Chen, Tumor microenvironment: Sanctuary of the devil, Cancer Letters, vol.368, issue.1, pp.7-13, 2015.
DOI : 10.1016/j.canlet.2015.07.039

M. Castells, B. Thibault, J. Delord, and B. Couderc, Implication of Tumor Microenvironment in Chemoresistance: Tumor-Associated Stromal Cells Protect Tumor Cells from Cell Death, International Journal of Molecular Sciences, vol.9, issue.3, pp.9545-9571, 2012.
DOI : 10.1016/j.ccr.2012.02.022

B. Thibault, M. Castells, J. Delord, and B. Couderc, Ovarian cancer microenvironment: implications for cancer dissemination and chemoresistance acquisition, Cancer and Metastasis Reviews, vol.13, issue.2, pp.17-39, 2014.
DOI : 10.1586/era.12.165

J. Hamanishi, Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer, Proceedings of the National Academy of Sciences, vol.17, issue.2, pp.3360-3365, 2007.
DOI : 10.1093/intimm/dxh194

E. Sato, Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer, Proceedings of the National Academy of Sciences, vol.34, issue.2, pp.18538-18543, 2005.
DOI : 10.1002/eji.200324181

C. Bokemeyer, A phase II study of catumaxomab administered intra- and postoperatively as part of a multimodal approach in primarily resectable gastric cancer, Gastric Cancer, vol.29, pp.833-842, 2015.
DOI : 10.1200/JCO.2010.34.3939

M. Goebeler and R. Bargou, Blinatumomab: a CD19/CD3 bispecific T cell engager (BiTE) with unique anti-tumor efficacy, Leukemia & Lymphoma, vol.124, issue.5, pp.1021-1032, 2016.
DOI : 10.1093/intimm/dxu089

J. Wu, J. Fu, M. Zhang, and D. Liu, Blinatumomab: a bispecific T cell engager (BiTE) antibody against CD19/CD3 for refractory acute lymphoid leukemia, Journal of Hematology & Oncology, vol.124, issue.21, p.104, 2015.
DOI : 10.1126/scitranslmed.3005930

URL : http://doi.org/10.1186/s13045-015-0195-4

D. Palma, M. Lewis, and C. E. , Macrophage regulation of tumor responses to anticancer therapies, Cancer Cell, vol.23, pp.277-286, 2013.

J. W. Pollard, Opinion: Tumour-educated macrophages promote tumour progression and metastasis, Nature Reviews Cancer, vol.4, issue.1, pp.71-78, 2004.
DOI : 10.1038/nrc1256

M. Castells, Ovarian ascites-derived Hospicells promote angiogenesis via activation of macrophages, Cancer Letters, vol.326, issue.1, pp.59-68, 2012.
DOI : 10.1016/j.canlet.2012.07.020

C. Guo, A. Buranych, D. Sarkar, P. B. Fisher, and X. Wang, The role of tumor-associated macrophages in tumor vascularization, Vascular Cell, vol.5, issue.1, p.20, 2013.
DOI : 10.1186/2045-824X-5-20

S. Gordon, Alternative activation of macrophages, Nature Reviews Immunology, vol.3, issue.1, pp.23-35, 2003.
DOI : 10.1038/nri978

URL : https://hal.archives-ouvertes.fr/hal-00474829

P. Chen and P. Bonaldo, Role of Macrophage Polarization in Tumor Angiogenesis and Vessel Normalization, Int. Rev. Cell Mol. Biol, vol.301, pp.1-35, 2013.
DOI : 10.1016/B978-0-12-407704-1.00001-4

D. Duluc, Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells, Blood, vol.110, issue.13, pp.4319-4330, 2007.
DOI : 10.1182/blood-2007-02-072587

S. Nath and P. Mukherjee, MUC1: a multifaceted oncoprotein with a key role in cancer progression, Trends in Molecular Medicine, vol.20, issue.6, pp.332-342, 2014.
DOI : 10.1016/j.molmed.2014.02.007

E. Arriola and C. Ottensmeier, TG4010: a vaccine with a therapeutic role in cancer, Immunotherapy, vol.8, issue.5, pp.511-519, 2016.
DOI : 10.2217/imt-2016-0015

V. Kotla, Mechanism of action of lenalidomide in hematological malignancies, Journal of Hematology & Oncology, vol.2, issue.1
DOI : 10.1186/1756-8722-2-36

X. Leleu, Thalidomide: mechanisms of action and new insights in hematology

G. Fouquet, Pomalidomide for multiple myeloma, Expert Review of Hematology, vol.7, issue.6, pp.613-618, 2015.
DOI : 10.1586/17474086.2014.966074

URL : http://citeseerx.ist.psu.edu/viewdoc/summary?doi=10.1.1.653.6228

D. Leclers, Lymphatic vessels and cancer, Med. Sci. MS, vol.21, pp.839-847, 2005.

W. R. Wilson and M. P. Hay, Targeting hypoxia in cancer therapy, Nature Reviews Cancer, vol.13, issue.6, pp.393-410, 2011.
DOI : 10.1158/1078-0432.CCR-06-2126

R. K. Jain, Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy, Science, vol.307, issue.5706, pp.58-62, 2005.
DOI : 10.1126/science.1104819

M. Sefidgar, Numerical modeling of drug delivery in a dynamic solid tumor microvasculature, Microvascular Research, vol.99, pp.43-56, 2015.
DOI : 10.1016/j.mvr.2015.02.007

J. Cosse and C. Michiels, Tumour Hypoxia Affects the Responsiveness of Cancer Cells to Chemotherapy and Promotes Cancer Progression, Anti-Cancer Agents in Medicinal Chemistry, vol.8, issue.7, pp.790-797, 2008.
DOI : 10.2174/187152008785914798

A. Régina, Multidrug resistance in brain tumors: roles of the blood-brain barrier, Cancer and Metastasis Reviews, vol.20, issue.1/2, pp.13-25, 2001.
DOI : 10.1023/A:1013104423154

R. S. Huang and M. J. Ratain, Pharmacogenetics and pharmacogenomics of anticancer agents, CA: A Cancer Journal for Clinicians, vol.59, issue.1, pp.42-55, 2009.
DOI : 10.3322/caac.20002

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3109906

J. Wang and Y. Huang, Pharmacogenomics of Sex Difference in Chemotherapeutic Toxicity, Current Drug Discovery Technologies, vol.4, issue.1, pp.59-68, 2007.
DOI : 10.2174/157016307781115485

G. Milano and A. Chamorey, Clinical pharmacokinetics of 5-fluorouracil with consideration of chronopharmacokinetics, Chronobiology International, vol.86, issue.1, pp.177-189, 2002.
DOI : 10.1093/jnci/86.21.1608

L. Galluzzi, Molecular mechanisms of cisplatin resistance, Oncogene, vol.12, issue.15, pp.1869-1883, 2012.
DOI : 10.1038/35060032

E. Chatelut, P. Canal, and R. Bugat, Pharmacokinetics and individual dose adjustment of carboplatin], Spec No, pp.17-23, 2000.

C. Holohan, S. Van-schaeybroeck, D. B. Longley, and P. G. Johnston, Cancer drug resistance: an evolving paradigm, Nature Reviews Cancer, vol.14, issue.10, pp.714-726, 2013.
DOI : 10.1093/neuonc/nos158

P. Sève, T. Reiman, and C. Dumontet, The role of ??III tubulin in predicting chemoresistance in non-small cell lung cancer, Lung Cancer, vol.67, issue.2, pp.136-143, 2010.
DOI : 10.1016/j.lungcan.2009.09.007

A. Keating, Mesenchymal Stromal Cells: New Directions, Cell Stem Cell, vol.10, issue.6, pp.709-716, 2012.
DOI : 10.1016/j.stem.2012.05.015

URL : http://doi.org/10.1016/j.stem.2012.06.004

M. Dominici, Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement, Cytotherapy, vol.8, issue.4, pp.315-317, 2006.
DOI : 10.1080/14653240600855905

J. S. Mohal, H. D. Tailor, and W. S. Khan, Sources of Adult Mesenchymal Stem Cells and their Applicability for Musculoskeletal Applications, Current Stem Cell Research & Therapy, vol.7, issue.2, pp.103-109, 2012.
DOI : 10.2174/157488812799219027

Q. He, C. Wan, and G. Li, Concise Review: Multipotent Mesenchymal Stromal Cells in Blood, STEM CELLS, vol.86, issue.(suppl), pp.69-77, 2007.
DOI : 10.1091/mbc.E02-02-0105

S. Boeuf and W. Richter, Chondrogenesis of mesenchymal stem cells: role of tissue source and inducing factors, Stem Cell Research & Therapy, vol.1, issue.4, p.31, 2010.
DOI : 10.1186/scrt31

F. Rastegar, Mesenchymal stem cells: Molecular characteristics and clinical applications, World J. Stem Cells, vol.2, pp.67-80, 2010.

G. Chen, In??vivo real-time visualization of mesenchymal stem cells tropism for cutaneous regeneration using NIR-II fluorescence imaging, Biomaterials, vol.53, pp.265-273, 2015.
DOI : 10.1016/j.biomaterials.2015.02.090

M. F. Pittenger, Multilineage Potential of Adult Human Mesenchymal Stem Cells, Science, vol.284, issue.5411, pp.143-147, 1999.
DOI : 10.1126/science.284.5411.143

Y. Jiang, Pluripotency of mesenchymal stem cells derived from adult marrow, Nature, vol.92, issue.6893, pp.41-49, 2002.
DOI : 10.1038/72753

A. M. Hocking and N. S. Gibran, Mesenchymal stem cells: Paracrine signaling and differentiation during cutaneous wound repair, Experimental Cell Research, vol.316, issue.14, pp.2213-2219, 2010.
DOI : 10.1016/j.yexcr.2010.05.009

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2902653

J. Dittmer and B. Leyh, Paracrine effects of stem cells in wound healing and cancer progression (Review), International Journal of Oncology, vol.44, pp.1789-1798, 2014.
DOI : 10.3892/ijo.2014.2385

J. M. Santos, Three-dimensional spheroid cell culture of umbilical cord tissue-derived mesenchymal stromal cells leads to enhanced paracrine induction of wound healing, Stem Cell Research & Therapy, vol.56, issue.1, p.90, 2015.
DOI : 10.1124/pr.56.4.3

URL : https://hal.archives-ouvertes.fr/pasteur-01164725

K. M. Akram, S. Samad, M. A. Spiteri, and N. R. Forsyth, Mesenchymal stem cells promote alveolar epithelial cell wound repair in vitro through distinct migratory and paracrine mechanisms, Respiratory Research, vol.14, issue.1, p.9, 2013.
DOI : 10.1186/1465-9921-14-9

URL : http://doi.org/10.1186/1465-9921-14-9

C. Touboul, F. Vidal, J. Pasquier, R. Lis, and A. Rafii, Role of mesenchymal cells in the natural history of ovarian cancer: a review, Journal of Translational Medicine, vol.36, issue.9737, p.271, 2014.
DOI : 10.1016/j.ejso.2010.04.005

URL : https://hal.archives-ouvertes.fr/inserm-01202727

H. Li and X. Fu, Mechanisms of action of mesenchymal stem cells in cutaneous wound repair and regeneration, Cell and Tissue Research, vol.10, issue.3, pp.371-377, 2012.
DOI : 10.1517/14712590903456011

L. Chen, E. E. Tredget, P. Y. Wu, and Y. Wu, Paracrine Factors of Mesenchymal Stem Cells Recruit Macrophages and Endothelial Lineage Cells and Enhance Wound Healing, PLoS ONE, vol.100, issue.4704, p.1886, 2008.
DOI : 10.1371/journal.pone.0001886.t003

L. Martinet, Hospicells derived from ovarian cancer stroma inhibit T-cell immune responses, International Journal of Cancer, vol.13, pp.2143-2152, 2010.
DOI : 10.4049/jimmunol.168.2.689

L. Martinet, A regulatory cross-talk between V??9V??2 T lymphocytes and mesenchymal stem cells, European Journal of Immunology, vol.168, issue.3, pp.752-762, 2009.
DOI : 10.4049/jimmunol.168.12.6336

R. Taran, In vitro and in vivo neurogenic potential of mesenchymal stem cells isolated from different sources, Journal of Biosciences, vol.30, issue.1, pp.157-169, 2014.
DOI : 10.1016/j.ijdevneu.2012.05.006

N. Joyce, Mesenchymal stem cells for the treatment of neurodegenerative disease, Regenerative Medicine, vol.5, issue.6, pp.933-946, 2010.
DOI : 10.2217/rme.10.72

F. G. Teixeira, M. M. Carvalho, N. Sousa, and A. J. Salgado, Mesenchymal stem cells secretome: a new paradigm for central nervous system regeneration?, Cellular and Molecular Life Sciences, vol.7, issue.4, pp.3871-3882, 2013.
DOI : 10.1016/j.stem.2010.09.009

C. M. Lewis and M. Suzuki, Therapeutic applications of mesenchymal stem cells for amyotrophic lateral sclerosis, Stem Cell Research & Therapy, vol.5, issue.2, p.32, 2014.
DOI : 10.1186/scrt421

L. Ferroni, Potential for neural differentiation of mesenchymal stem cells Adv

O. Sadan, E. Melamed, and D. Offen, Bone-marrow-derived mesenchymal stem cell therapy for neurodegenerative diseases, Expert Opinion on Biological Therapy, vol.65, issue.12, pp.1487-1497, 2009.
DOI : 10.1517/14712590903186956

S. Frausin, Wharton's jelly derived mesenchymal stromal cells: Biological properties, induction of neuronal phenotype and current applications in neurodegeneration research, Acta Histochemica, vol.117, issue.4-5, pp.329-338, 2015.
DOI : 10.1016/j.acthis.2015.02.005

J. S. Park, S. Suryaprakash, Y. Lao, and K. W. Leong, Engineering mesenchymal stem cells for regenerative medicine and drug delivery, Methods, vol.84, pp.3-16, 2015.
DOI : 10.1016/j.ymeth.2015.03.002

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4526354

M. B. Murphy, K. Moncivais, and A. Caplan, Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine, Experimental & Molecular Medicine, vol.154, issue.11, p.54, 2013.
DOI : 10.3324/haematol.11869

URL : http://www.nature.com/emm/journal/v45/n11/pdf/emm201394a.pdf

E. Spaeth, A. Klopp, J. Dembinski, M. Andreeff, and F. Marini, Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells, Gene Therapy, vol.285, issue.10, pp.730-738, 2008.
DOI : 10.1634/stemcells.2004-0176

S. W. Olechnowicz and C. M. Edwards, Contributions of the Host Microenvironment to Cancer-Induced Bone Disease, Cancer Research, vol.74, issue.6, pp.1625-1631, 2014.
DOI : 10.1158/0008-5472.CAN-13-2645

R. N. Kaplan, VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche, Nature, vol.174, issue.7069, pp.820-827, 2005.
DOI : 10.4049/jimmunol.174.6.3686

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2945882

L. Kucerova and S. Skolekova, Tumor microenvironment and the role of mesenchymal stromal cells, Neoplasma, vol.60, issue.01, pp.1-10, 2013.
DOI : 10.4149/neo_2013_001

P. Barcellos-de-souza, V. Gori, F. Bambi, and P. Chiarugi, Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, vol.1836, issue.2, pp.321-335, 2013.
DOI : 10.1016/j.bbcan.2013.10.004

S. Kidd, Direct Evidence of Mesenchymal Stem Cell Tropism for Tumor and Wounding Microenvironments Using In Vivo Bioluminescent Imaging, Stem Cells, vol.116, issue.10, pp.2614-2623, 2009.
DOI : 10.1056/NEJM198612253152606

R. Carrero, IL1?? Induces Mesenchymal Stem Cells Migration and Leucocyte Chemotaxis Through NF-??B, Stem Cell Reviews and Reports, vol.9, issue.Suppl 1, pp.905-916, 2012.
DOI : 10.3727/096368910X543376

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3412085

X. Liu, SDF-1/CXCR4 axis modulates bone marrow mesenchymal stem cell apoptosis, migration and cytokine secretion, Protein & Cell, vol.21, issue.10, pp.845-854, 2011.
DOI : 10.1096/fj.06-6558com

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4875294

J. M. Houthuijzen, L. G. Daenen, J. M. Roodhart, and E. E. Voest, The role of mesenchymal stem cells in anti-cancer drug resistance and tumour progression, British Journal of Cancer, vol.29, issue.12, pp.1901-1906, 2012.
DOI : 10.1016/0163-7827(90)90005-6

J. M. Roodhart, Mesenchymal Stem Cells Induce Resistance to Chemotherapy through the Release of Platinum-Induced Fatty Acids, Cancer Cell, vol.20, issue.3, pp.370-383, 2011.
DOI : 10.1016/j.ccr.2011.08.010

A. E. Karnoub, Mesenchymal stem cells within tumour stroma promote breast cancer metastasis, Nature, vol.9, issue.7162, pp.557-563, 2007.
DOI : 10.3233/BD-2007-26107

R. Lis, Mesenchymal cell interaction with ovarian cancer cells induces a background dependent pro-metastatic transcriptomic profile, Journal of Translational Medicine, vol.12, issue.1, p.59, 2014.
DOI : 10.1172/JCI45273

URL : http://doi.org/10.1186/1479-5876-12-59

M. B. Meads, L. A. Hazlehurst, and W. S. Dalton, The Bone Marrow Microenvironment as a Tumor Sanctuary and Contributor to Drug Resistance, Clinical Cancer Research, vol.14, issue.9, pp.2519-2526, 2008.
DOI : 10.1158/1078-0432.CCR-07-2223

A. Rafii, Oncologic Trogocytosis of an Original Stromal Cells Induces Chemoresistance of Ovarian Tumours, PLoS ONE, vol.171, issue.2, p.3894, 2008.
DOI : 10.1371/journal.pone.0003894.s002

M. Pasquet, Hospicells (ascites-derived stromal cells) promote tumorigenicity and angiogenesis, International Journal of Cancer, vol.16, pp.2090-2101, 2010.
DOI : 10.1016/S0002-9440(10)65669-6

M. Castells, Microenvironment mesenchymal cells protect ovarian cancer cell lines from apoptosis by inhibiting XIAP inactivation, Cell Death and Disease, vol.126, issue.10, p.887, 2013.
DOI : 10.1002/pmic.200700231

URL : http://doi.org/10.1038/cddis.2013.384

L. Bochet, Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer Adipose-derived stromal cells inhibit prostate cancer cell proliferation inducing apoptosis, Cancer Res. Biochem. Biophys. Res. Commun, vol.73, issue.446, pp.5657-5668, 2013.

L. P. Mueller, Presence of Mesenchymal Stem Cells in Human Bone Marrow After Exposure to Chemotherapy: Evidence of Resistance to Apoptosis Induction, Stem Cells, vol.27, issue.12, pp.2753-2765, 2006.
DOI : 10.1172/JCI0216098

O. S. Beane, V. C. Fonseca, and E. M. Darling, Adipose-derived stem cells retain their regenerative potential after methotrexate treatment, Experimental Cell Research, vol.327, issue.2, pp.222-233, 2014.
DOI : 10.1016/j.yexcr.2014.06.015

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4164584

J. A. Burger and T. J. Kipps, Chemokine Receptors and Stromal Cells in the Homing and Homeostasis of Chronic Lymphocytic Leukemia B Cells, Leukemia & Lymphoma, vol.56, issue.3, pp.461-466, 2002.
DOI : 10.1002/1097-0142(19850715)56:2<328::AID-CNCR2820560221>3.0.CO;2-Q

M. B. Meads, R. A. Gatenby, and W. S. Dalton, Environment-mediated drug resistance: a major contributor to minimal residual disease, Nature Reviews Cancer, vol.112, issue.9, pp.665-674, 2009.
DOI : 10.1038/nrm1229

A. V. Kurtova, Diverse marrow stromal cells protect CLL cells from spontaneous and drug-induced apoptosis: development of a reliable and reproducible system to assess stromal cell adhesion-mediated drug resistance, Blood, vol.114, issue.20, pp.4441-4450, 2009.
DOI : 10.1182/blood-2009-07-233718

N. N. Bewry, Stat3 contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment model of drug resistance, Molecular Cancer Therapeutics, vol.7, issue.10, pp.3169-3175, 2008.
DOI : 10.1158/1535-7163.MCT-08-0314

K. Brocke-heidrich, Interleukin-6-dependent gene expression profiles in multiple myeloma INA-6 cells reveal a Bcl-2 family-independent survival pathway closely associated with Stat3 activation, Blood, vol.103, issue.1, pp.242-251, 2004.
DOI : 10.1182/blood-2003-04-1048

Y. Yamagiwa, C. Marienfeld, F. Meng, M. Holcik, and T. Patel, Translational Regulation of X-Linked Inhibitor of Apoptosis Protein by Interleukin-6: A Novel Mechanism of Tumor Cell Survival, Cancer Research, vol.64, issue.4, pp.1293-1298, 2004.
DOI : 10.1158/0008-5472.CAN-03-2517

J. Moreaux, BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone, Blood, vol.103, issue.8, pp.3148-3157, 2004.
DOI : 10.1182/blood-2003-06-1984

URL : https://hal.archives-ouvertes.fr/inserm-00129502

L. Jin, CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells, Molecular Cancer Therapeutics, vol.7, issue.1, pp.48-58, 2008.
DOI : 10.1158/1535-7163.MCT-07-0042

R. Ji, Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer, Cell Cycle, vol.1, issue.4, pp.2473-2483, 2015.
DOI : 10.3402/jev.v3.24783

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4613597

R. Lis, Mesenchymal Cell Interaction with Ovarian Cancer Cells Triggers Pro-Metastatic Properties, PLoS ONE, vol.474, issue.5, p.38340, 2012.
DOI : 10.1371/journal.pone.0038340.s002

URL : http://doi.org/10.1371/journal.pone.0038340

M. Gupta, Hematopoietic cells from Gadd45a- and Gadd45b-deficient mice are sensitized to genotoxic-stress-induced apoptosis, Oncogene, vol.18, issue.48, pp.7170-7179, 2005.
DOI : 10.1038/sj.onc.1202885

D. Smaele and E. , Induction of gadd45beta by NF-kappaB downregulates proapoptotic JNK signalling, Nature, vol.414, issue.6861, pp.308-313, 2001.
DOI : 10.1038/35104560

C. Riganti, J. Kopecka, E. Panada, S. Barak, and M. Rubinstein, The Role of C/EBP-?? LIP in Multidrug Resistance, JNCI: Journal of the National Cancer Institute, vol.107, issue.5, 2015.
DOI : 10.1093/jnci/djv046

S. Pillozzi, Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers, Blood, vol.117, issue.3, pp.902-914, 2011.
DOI : 10.1182/blood-2010-01-262691

M. Mraz, Bone marrow stromal cells protect lymphoma B-cells from rituximab-induced apoptosis and targeting integrin ??-4-??-1 (VLA-4) with natalizumab can overcome this resistance, British Journal of Haematology, vol.69, issue.1, pp.53-64, 2011.
DOI : 10.1158/0008-5472.CAN-08-4173

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4405035

J. Xu, P. Zhou, W. Wang, A. Sun, and F. Guo, RelB, together with RelA, sustains cell survival and confers proteasome inhibitor sensitivity of chronic lymphocytic leukemia cells from bone marrow, Journal of Molecular Medicine, vol.117, issue.1, pp.77-92, 2014.
DOI : 10.1182/blood-2010-07-155663

Y. Yang, Wnt pathway contributes to the protection by bone marrow stromal cells of acute lymphoblastic leukemia cells and is a potential therapeutic target, Cancer Letters, vol.333, issue.1, pp.9-17, 2013.
DOI : 10.1016/j.canlet.2012.11.056

B. Zhang, Microenvironmental protection of CML stem and progenitor cells from tyrosine kinase inhibitors through N-cadherin and Wnt-??-catenin signaling, Blood, vol.121, issue.10, pp.1824-1838, 2013.
DOI : 10.1182/blood-2012-02-412890

A. Scherzed, BMSC enhance the survival of paclitaxel treated squamous cell carcinoma cells in vitro, Cancer Biology & Therapy, vol.11, issue.3, pp.349-357, 2011.
DOI : 10.4161/cbt.11.3.14179

L. Borriello and Y. A. Declerck, Le microenvironnement tumoral et la r??sistance th??rapeutique, m??decine/sciences, vol.30, issue.4, pp.445-451, 2014.
DOI : 10.1051/medsci/20143004021

F. Sanz-rodríguez, A. Hidalgo, and J. Teixidó, Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-mediated multiple myeloma cell adhesion to CS-1/fibronectin and VCAM-1, Blood, vol.97, issue.2, pp.346-351, 2001.
DOI : 10.1182/blood.V97.2.346

N. Yanamandra, Tipifarnib and Bortezomib Are Synergistic and Overcome Cell Adhesion-Mediated Drug Resistance in Multiple Myeloma and Acute Myeloid Leukemia, Clinical Cancer Research, vol.12, issue.2, pp.591-599, 2006.
DOI : 10.1158/1078-0432.CCR-05-1792

Y. Dai, T. S. Lawrence, and L. Xu, Overcoming cancer therapy resistance by targeting inhibitors of apoptosis proteins and nuclear factor-kappa B, Am. J. Transl. Res, vol.1, pp.1-15, 2009.

J. W. Antoon, Targeting NF??B mediated breast cancer chemoresistance through selective inhibition of sphingosine kinase-2, Cancer Biology & Therapy, vol.58, issue.7, pp.678-689, 2011.
DOI : 10.1158/1078-0432.CCR-06-1426

J. Lundqvist, C. W. Yde, and A. Lykkesfeldt, 1??,25-Dihydroxyvitamin D3 inhibits cell growth and NF??B signaling in tamoxifen-resistant breast cancer cells, Steroids, vol.85, issue.85, pp.30-35, 2014.
DOI : 10.1016/j.steroids.2014.04.001

D. Cilloni, The NF-??B pathway blockade by the IKK inhibitor PS1145 can overcome Imatinib resistance, Leukemia, vol.12, issue.1, pp.61-67, 2006.
DOI : 10.1016/S0065-230X(02)85003-5

H. Aberle, A. Bauer, J. Stappert, A. Kispert, and R. Kemler, ??-catenin is a target for the ubiquitin???proteasome pathway, The EMBO Journal, vol.16, issue.13, pp.3797-3804, 1997.
DOI : 10.1093/emboj/16.13.3797

J. Tian, H. He, and G. Lei, Wnt/??-catenin pathway in bone cancers, Tumor Biology, vol.239, issue.10, pp.9439-9445, 2014.
DOI : 10.1016/j.intimp.2013.11.021

M. Kahn, Can we safely target the WNT pathway?, Nature Reviews Drug Discovery, vol.31, issue.7, pp.513-532, 2014.
DOI : 10.1158/1078-0432.CCR-09-2943

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4426976

B. Xia, c-Myc plays part in drug resistance mediated by bone marrow stromal cells in acute myeloid leukemia, Leukemia Research, vol.39, issue.1, pp.92-99, 2015.
DOI : 10.1016/j.leukres.2014.11.004

B. E. Kadera, MicroRNA-21 in Pancreatic Ductal Adenocarcinoma Tumor-Associated Fibroblasts Promotes Metastasis, PLoS ONE, vol.327, issue.8, p.71978, 2013.
DOI : 10.1371/journal.pone.0071978.s006

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3750050/pdf

B. G. Cuiffo and A. E. Karnoub, Mesenchymal stem cells in tumor development, Cell Adhesion & Migration, vol.61, issue.3, pp.220-230, 2012.
DOI : 10.1126/science.1099513

B. I. Koh and Y. Kang, The pro-metastatic role of bone marrow-derived cells: a focus on MSCs and regulatory T cells, EMBO reports, vol.30, issue.5, pp.412-422, 2012.
DOI : 10.1007/s10555-011-9280-5

S. A. Bergfeld and Y. A. Declerck, Bone marrow-derived mesenchymal stem cells and the tumor microenvironment, Cancer and Metastasis Reviews, vol.15, issue.289???98, pp.249-261, 2010.
DOI : 10.3109/10428199809059251

L. Iacono and M. , ATF2 contributes to cisplatin resistance in non-small cell lung cancer and celastrol induces cisplatin resensitization through inhibition of JNK/ATF2 pathway, International Journal of Cancer, vol.273, issue.11, pp.2598-2609, 2015.
DOI : 10.1074/jbc.273.9.4928

E. Lau, Transcriptional repression of IFN??1 by ATF2 confers melanoma resistance to therapy, Oncogene, vol.136, issue.46, 2015.
DOI : 10.1002/ijc.29004

Z. Ronai, ATF2 confers radiation resistance to human melanoma cells, Oncogene, vol.16, issue.4, pp.523-531, 1998.
DOI : 10.1038/sj.onc.1201566

J. O. Lauchle, Response and resistance to MEK inhibition in leukaemias initiated by hyperactive Ras, Nature, vol.85, issue.7262, pp.411-414, 2009.
DOI : 10.1038/nature08279

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4119783

M. Takahashi, Loss of B-cell translocation gene 2 expression in estrogen receptor-positive breast cancer predicts tamoxifen resistance, Cancer Science, vol.366, issue.Suppl. 2, pp.675-682, 2014.
DOI : 10.1056/NEJMoa1109653

Y. Nefedova, T. H. Landowski, and W. S. Dalton, Bone marrow stromal-derived soluble factors and direct cell contact contribute to de novo drug resistance of myeloma cells by distinct mechanisms, Leukemia, vol.17, issue.6, pp.1175-1182, 2003.
DOI : 10.1038/sj.leu.2402924

T. Lwin, Cell adhesion induces p27Kip1-associated cell-cycle arrest through down-regulation of the SCFSkp2 ubiquitin ligase pathway in mantle-cell and other non-Hodgkin B-cell lymphomas, Blood, vol.110, issue.5, pp.1631-1638, 2007.
DOI : 10.1182/blood-2006-11-060350

Z. Wei, Bone marrow mesenchymal stem cells from leukemia patients inhibit growth and apoptosis in serum-deprived K562 cells, Journal of Experimental & Clinical Cancer Research, vol.28, issue.1, p.141, 2009.
DOI : 10.1186/1756-9966-28-141

URL : http://doi.org/10.1186/1756-9966-28-141

R. Ramasamy, Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth, Leukemia, vol.102, issue.2, pp.304-310, 2007.
DOI : 10.1038/sj.leu.2404489

Y. Nefedova, P. Cheng, M. Alsina, W. S. Dalton, and D. I. Gabrilovich, Involvement of Notch-1 signaling in bone marrow stroma-mediated de novo drug resistance of myeloma and other malignant lymphoid cell lines, Blood, vol.103, issue.9, pp.3503-3510, 2004.
DOI : 10.1182/blood-2003-07-2340

N. Kamdje and A. H. , Notch-3 and Notch-4 signaling rescue from apoptosis human B-ALL cells in contact with human bone marrow-derived mesenchymal stromal cells, Blood, vol.118, issue.2, pp.380-389, 2011.
DOI : 10.1182/blood-2010-12-326694

N. Kamdje and A. H. , Role of stromal cell-mediated Notch signaling in CLL resistance to chemotherapy, Blood Cancer Journal, vol.25, issue.5, p.73, 2012.
DOI : 10.1038/leu.2011.103

Z. Zeng, Inhibition of CXCR4 with the novel RCP168 peptide overcomes stroma-mediated chemoresistance in chronic and acute leukemias, Molecular Cancer Therapeutics, vol.5, issue.12, pp.3113-3121, 2006.
DOI : 10.1158/1535-7163.MCT-06-0228

K. E. Corcoran, Mesenchymal Stem Cells in Early Entry of Breast Cancer into Bone Marrow, PLoS ONE, vol.285, issue.6, p.2563, 2008.
DOI : 10.1371/journal.pone.0002563.s005

F. Vianello, Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis, Haematologica, vol.95, issue.7, pp.1081-1089, 2010.
DOI : 10.3324/haematol.2009.017178

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2895031

R. Lis, Tumor associated mesenchymal stem cells protects ovarian cancer cells from hyperthermia through CXCL12, International Journal of Cancer, vol.2010, issue.3, pp.715-725, 2011.
DOI : 10.1111/IGC.0b013e3181b7a40e

J. Kim, Microenvironmental Remodeling as a Parameter and Prognostic Factor of Heterogeneous Leukemogenesis in Acute Myelogenous Leukemia, Cancer Research, vol.75, issue.11, pp.2222-2231, 2015.
DOI : 10.1158/0008-5472.CAN-14-3379

A. Peled, Cells from CML Patients Have Lower Integrin-Dependent Migration and Adhesion in Response to the Chemokine SDF-1, Stem Cells, vol.27, issue.3, pp.259-266, 2002.
DOI : 10.1155/1998/764986

K. Kojima, p53 activation of mesenchymal stromal cells partially abrogates microenvironment-mediated resistance to FLT3 inhibition in AML through HIF-1??-mediated down-regulation of CXCL12, Blood, vol.118, issue.16, pp.4431-4439, 2011.
DOI : 10.1182/blood-2011-02-334136

F. Hubin, Murine Bone Marrow Stromal Cells Sustain In Vivo the Survival of Hematopoietic Stem Cells and the Granulopoietic Differentiation of More Mature Progenitors, Stem Cells, vol.76, issue.10, pp.1626-1633, 2005.
DOI : 10.1634/stemcells.2005-0041

J. Hoellenriegel, The Spiegelmer NOX-A12, a novel CXCL12 inhibitor, interferes with chronic lymphocytic leukemia cell motility and causes chemosensitization, Blood, vol.123, issue.7, pp.1032-1039, 2014.
DOI : 10.1182/blood-2013-03-493924

S. J. Greco, AMD3100-mediated production of interleukin-1 from mesenchymal stem cells is key to chemosensitivity of breast cancer cells, Am. J. Cancer Res, vol.1, pp.701-715, 2011.

J. Rossi, Z. Lu, M. Jourdan, and B. Klein, Interleukin-6 as a Therapeutic Target, Clinical Cancer Research, vol.21, issue.6, pp.1248-1257, 2015.
DOI : 10.1158/1078-0432.CCR-14-2291

A. Lichtenstein, Y. Tu, C. Fady, R. Vescio, and J. Berenson, Interleukin-6 Inhibits Apoptosis of Malignant Plasma Cells, Cellular Immunology, vol.162, issue.2, pp.248-255, 1995.
DOI : 10.1006/cimm.1995.1076

Z. Duan, Signal Transducers and Activators of Transcription 3 Pathway Activation in Drug-Resistant Ovarian Cancer, Clinical Cancer Research, vol.12, issue.17, pp.5055-5063, 2006.
DOI : 10.1158/1078-0432.CCR-06-0861

A. Dreuw, Interleukin-6-Type Cytokines Upregulate Expression of Multidrug Resistance-Associated Proteins in NHEK and Dermal Fibroblasts, Journal of Investigative Dermatology, vol.124, issue.1, pp.28-37, 2005.
DOI : 10.1111/j.0022-202X.2004.23499.x

K. H. Shain, ??1 Integrin Adhesion Enhances IL-6-Mediated STAT3 Signaling in Myeloma Cells: Implications for Microenvironment Influence on Tumor Survival and Proliferation, Cancer Research, vol.69, issue.3, pp.1009-1015, 2009.
DOI : 10.1158/0008-5472.CAN-08-2419

Y. Wang, Autocrine production of interleukin-6 confers cisplatin and paclitaxel resistance in ovarian cancer cells, Cancer Letters, vol.295, issue.1, pp.110-123, 2010.
DOI : 10.1016/j.canlet.2010.02.019

Y. Wang, Autocrine production of interleukin-6 confers ovarian cancer cells resistance to tamoxifen via ER isoforms and SRC-1, Molecular and Cellular Endocrinology, vol.382, issue.2, pp.791-803, 2014.
DOI : 10.1016/j.mce.2013.10.029

T. Efferth, U. Fabry, and R. Osieka, Interleukin-6 affects melphalan-induced DNA damage and repair in human multiple myeloma cells, Anticancer Res, vol.22, pp.231-234, 2002.

J. Corre, Bone marrow mesenchymal stem cells are abnormal in multiple myeloma, Leukemia, vol.63, pp.1079-1088, 2007.
DOI : 10.1016/S0002-9440(10)63348-2

URL : https://hal.archives-ouvertes.fr/inserm-00270565

J. Golay, The histone deacetylase inhibitor ITF2357 has anti-leukemic activity in vitro and in vivo and inhibits IL-6 and VEGF production by stromal cells, Leukemia, vol.132, issue.9, pp.1892-1900, 2007.
DOI : 10.1038/sj.leu.2404282

B. Vanhaesebroeck, J. Guillermet-guibert, M. Graupera, and B. Bilanges, The emerging mechanisms of isoform-specific PI3K signalling, Nature Reviews Molecular Cell Biology, vol.11, issue.5, pp.329-341, 2010.
DOI : 10.1128/MCB.10.12.6742

L. M. Thorpe, H. Yuzugullu, and J. J. Zhao, PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting, Nature Reviews Cancer, vol.26, issue.1, pp.7-24, 2015.
DOI : 10.1016/j.ccr.2014.05.020

X. Sheng and S. D. Mittelman, The Role of Adipose Tissue and Obesity in Causing Treatment Resistance of Acute Lymphoblastic Leukemia, Frontiers in Pediatrics, vol.7, issue.4, p.53, 2014.
DOI : 10.1371/journal.pone.0033738

S. Tung, PPAR?? and fatty acid oxidation mediate glucocorticoid resistance in chronic lymphocytic leukemia, Blood, vol.122, issue.6, pp.969-980, 2013.
DOI : 10.1182/blood-2013-03-489468

A. V. Kurtova, Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance, Nature, vol.9, issue.7533, pp.209-213, 2015.
DOI : 10.1038/nrc1697

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4465385

S. P. Selvam and B. Ogretmen, Sphingosine Kinase/Sphingosine 1-Phosphate Signaling in Cancer Therapeutics and Drug Resistance, Handb. Exp. Pharmacol, pp.3-27978, 2013.
DOI : 10.1007/978-3-7091-1511-4_1

J. Corre, Bioactivity and Prognostic Significance of Growth Differentiation Factor GDF15 Secreted by Bone Marrow Mesenchymal Stem Cells in Multiple Myeloma, Cancer Research, vol.72, issue.6, pp.1395-1406, 2012.
DOI : 10.1158/0008-5472.CAN-11-0188

M. M. Keenan and J. Chi, Alternative Fuels for Cancer Cells, The Cancer Journal, vol.21, issue.2, pp.49-55, 2015.
DOI : 10.1097/PPO.0000000000000104

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4380238

E. A. Ehsanipour, Adipocytes Cause Leukemia Cell Resistance to L-Asparaginase via Release of Glutamine, Cancer Research, vol.73, issue.10, pp.2998-3006, 2013.
DOI : 10.1158/0008-5472.CAN-12-4402

S. Iwamoto, K. Mihara, J. R. Downing, C. Pui, and D. Campana, Mesenchymal cells regulate the response of acute lymphoblastic leukemia cells to asparaginase, Journal of Clinical Investigation, vol.117, issue.4, pp.1049-1057, 2007.
DOI : 10.1172/JCI30235

B. Yu, X. Zhang, and X. Li, Exosomes Derived from Mesenchymal Stem Cells, International Journal of Molecular Sciences, vol.12, issue.3
DOI : 10.1371/journal.pone.0082949

J. Lee, Exosomes Derived from Mesenchymal Stem Cells Suppress Angiogenesis by Down-Regulating VEGF Expression in Breast Cancer Cells, PLoS ONE, vol.361, issue.12, p.84256, 2013.
DOI : 10.1371/journal.pone.0084256.g006

URL : http://doi.org/10.1371/journal.pone.0084256

M. Ono, Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells, Science Signaling, vol.105, issue.30, p.63, 2014.
DOI : 10.1073/pnas.0804549105

Y. Lin, Study on the bone marrow mesenchymal stem cells induced drug resistance in the U937 cells and its mechanism, Chin. Med. J. (Engl.), vol.119, pp.905-910, 2006.

M. Konopleva, Stromal cells prevent apoptosis of AML cells by up-regulation of anti-apoptotic proteins, Leukemia, vol.16, issue.9, pp.1713-1724, 2002.
DOI : 10.1038/sj.leu.2402608

K. Balakrishnan, Influence of bone marrow stromal microenvironment on forodesine-induced responses in CLL primary cells, Blood, vol.116, issue.7, pp.1083-1091, 2010.
DOI : 10.1182/blood-2009-10-246199

X. Pei, Circumvention of Mcl-1-Dependent Drug Resistance by Simultaneous Chk1 and MEK1/2 Inhibition in Human Multiple Myeloma Cells, PLoS ONE, vol.19, issue.3, p.89064, 2014.
DOI : 10.1371/journal.pone.0089064.s006

. Anonyme, Guide affection de longue durée 30 'cancer de l, 1980.

S. Domcke, R. Sinha, D. A. Levine, C. Sander, and N. Schultz, Evaluating cell lines as tumour models by comparison of genomic profiles, Nature Communications, vol.29, p.2126, 2013.
DOI : 10.1038/nbt.1754

L. Picaud, Evaluation of the effects of hyaluronic acid-carboxymethyl cellulose barrier on ovarian tumor progression, Journal of Ovarian Research, vol.7, issue.1, p.40, 2014.
DOI : 10.1016/j.jconrel.2011.06.031