M. Raab, K. Podar, I. Breitkreutz, P. Richardson, and K. Anderson, Multiple myeloma, The Lancet, vol.374, issue.9686, pp.324-339, 2009.
DOI : 10.1016/S0140-6736(09)60221-X

URL : https://hal.archives-ouvertes.fr/inserm-00130206

A. Mahindra, J. Laubach, N. Raje, N. Munshi, P. Richardson et al., Latest advances and current challenges in the treatment of multiple myeloma, Nature Reviews Clinical Oncology, vol.364, issue.3, pp.135-143, 2012.
DOI : 10.1038/nrclinonc.2012.15

S. Treon, G. Teoh, M. Urashima, A. Ogata, D. Chauhan et al., Anti-estrogens induce apoptosis of multiple myeloma cells, Blood, vol.92, issue.5, pp.1749-1457, 1998.

T. Otsuki, O. Yamada, J. Kurebayashi, T. Moriya, H. Sakaguchi et al., Estrogen receptors in human myeloma cells, Cancer Res, vol.60, issue.5, pp.1434-1441, 2000.

J. Gauduchon, F. Gouilleux, S. Maillard, V. Marsaud, J. Renoir et al., 4-Hydroxytamoxifen Inhibits Proliferation of Multiple Myeloma Cells In vitro through Down-Regulation of c-Myc, Up-Regulation of p27Kip1, and Modulation of Bcl-2 Family Members, Clinical Cancer Research, vol.11, issue.6, pp.2345-2354, 2005.
DOI : 10.1158/1078-0432.CCR-04-1668

S. Olivier, P. Close, E. Castermans, L. De-leval, S. Tabruyn et al., Raloxifene-Induced Myeloma Cell Apoptosis: A Study of Nuclear Factor-??B Inhibition and Gene Expression Signature, Molecular Pharmacology, vol.69, issue.5, pp.1615-1623, 2006.
DOI : 10.1124/mol.105.020479

J. Gauduchon, A. Seguin, V. Marsaud, D. Clay, J. Renoir et al., Pure antiestrogen-induced G1-arrest in myeloma cells results from the reduced kinase activity of cyclin D3/CDK6 complexes whereas apoptosis is mediated by endoplasmic reticulum-dependent caspases, International Journal of Cancer, vol.2, issue.9, pp.2130-2141, 2008.
DOI : 10.1002/ijc.23310

G. Urbinati, D. Audisio, V. Marsaud, V. Plassat, S. Arpicco et al., Therapeutic potential of 12 Identiication and pharmacological characterization of cholesterol-5,6-epoxide hydrolase as a target of tamoxifen and AEBS ligands, Proc Natl Acad Sci, vol.107, issue.33, pp.13520-13525, 2010.

S. Silvente-poirot and M. Poirot, Cholesterol epoxide hydrolase and cancer, Current Opinion in Pharmacology, vol.12, issue.6, pp.696-703, 2012.
DOI : 10.1016/j.coph.2012.07.007

URL : https://hal.archives-ouvertes.fr/inserm-00743463

P. De-medina, B. Payré, N. Boubekeur, J. Bertrand-michel, F. Tercé et al., Ligands of the antiestrogen-binding site induce active cell death and autophagy in human breast cancer cells through the modulation of cholesterol metabolism, Cell Death and Differentiation, vol.66, issue.10, pp.1372-1384, 2009.
DOI : 10.1074/jbc.275.2.992

URL : https://hal.archives-ouvertes.fr/inserm-00519149

B. Hoang, A. Benavides, Y. Shi, P. Frost, and A. Lichtenstein, Effect of autophagy on multiple myeloma cell viability, Molecular Cancer Therapeutics, vol.8, issue.7, pp.1974-1984, 2009.
DOI : 10.1158/1535-7163.MCT-08-1177

L. Galluzzi, I. Vitale, J. Abrams, E. Alnemri, E. Baehrecke et al., Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012, Cell Death and Differentiation, vol.94, issue.1, pp.107-120, 2012.
DOI : 10.1083/jcb.200712051

D. Klionsky, F. Abdalla, H. Abeliovich, R. Abraham, A. Acevedo-arozena et al., Guidelines for the use and interpretation of assays for monitoring autophagy, Autophagy, vol.4, issue.4, pp.445-544, 2012.
DOI : 10.4161/auto.6.4.12244

URL : https://hal.archives-ouvertes.fr/hal-01343085

G. Segala, P. De-medina, L. Iuliono, C. Zerbinati, M. Paillasse et al., 5,6-Epoxy-cholesterols contribute to the anticancer pharmacology of Tamoxifen in breast cancer cells, Biochemical Pharmacology, vol.86, issue.1, 2013.
DOI : 10.1016/j.bcp.2013.02.031

URL : https://hal.archives-ouvertes.fr/inserm-00822322

D. Cirstea, T. Hideshima, S. Rodig, L. Santo, S. Pozzi et al., Dual Inhibition of Akt/Mammalian Target of Rapamycin Pathway by Nanoparticle Albumin-Bound-Rapamycin and Perifosine Induces Antitumor Activity in Multiple Myeloma, Molecular Cancer Therapeutics, vol.9, issue.4, pp.963-975, 2010.
DOI : 10.1158/1535-7163.MCT-09-0763

C. Palacios, R. Martín-pérez, A. López-pérez, A. Pandiella, and A. López-rivas, Autophagy inhibition sensitizes multiple myeloma cells to 17-dimethylaminoethylamino-17-demethoxygeldanamycin-induced apoptosis, Leukemia Research, vol.34, issue.11, pp.1533-1538, 2010.
DOI : 10.1016/j.leukres.2010.07.002

A. Puissant, G. Robert, and P. Auberger, Targeting autophagy to fight hematopoietic malignancies, Cell Cycle, vol.9, issue.17, pp.3470-3478, 2010.
DOI : 10.4161/cc.9.17.13048

M. Cea, A. Cagnetta, M. Fulciniti, Y. Tai, T. Hideshima et al., Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase (ERK1/2) inhibition, Blood, vol.120, issue.17, pp.3519-3529, 2012.
DOI : 10.1182/blood-2012-03-416776

F. Pene, Y. Claessens, O. Muller, F. Viguié, P. Mayeux et al., Role of the phosphatidylinositol 3-kinase/Akt and mTOR/P70S6-kinase pathways in the proliferation and apoptosis in multiple myeloma, Oncogene, vol.21, issue.43, pp.6587-6597, 2002.
DOI : 10.1038/sj.onc.1205923

V. Jordan, Chemoprevention of breast cancer with selective oestrogen-receptor modulators, Nature Reviews Cancer, vol.313, issue.1, pp.46-53, 2007.
DOI : 10.1038/nrc2048

E. Musgrove and R. Sutherland, Biological determinants of endocrine resistance in breast cancer, Nature Reviews Cancer, vol.10, issue.9, pp.631-643, 2009.
DOI : 10.1038/nrc2713

J. Renoir, V. Marsaud, and G. Lazennec, Estrogen receptor signaling as a target for novel breast cancer therapeutics, Biochemical Pharmacology, vol.85, issue.4, pp.449-465, 2013.
DOI : 10.1016/j.bcp.2012.10.018

URL : https://hal.archives-ouvertes.fr/inserm-00758201

A. Michallet, P. Mondiere, M. Taillardet, Y. Leverrier, L. Genestier et al., Compromising the Unfolded Protein Response Induces Autophagy-Mediated Cell Death in Multiple Myeloma Cells, PLoS ONE, vol.6, issue.10, p.25820, 2011.
DOI : 10.1371/journal.pone.0025820.g007

Y. Zang, S. Thomas, E. Chan, C. Kirk, M. Freilino et al., The next generation proteasome inhibitors carfilzomib and oprozomib activate prosurvival autophagy via induction of the unfolded protein response and ATF4, Autophagy, vol.8, issue.12, pp.1873-1874, 2012.
DOI : 10.4161/auto.22185

M. Shanmugam, S. Mcbrayer, J. Qian, K. Raikoff, M. Avram et al., Targeting Glucose Consumption and Autophagy in Myeloma with the Novel Nucleoside Analogue 8-Aminoadenosine, Journal of Biological Chemistry, vol.284, issue.39, pp.26816-26830, 2009.
DOI : 10.1074/jbc.M109.000646

Y. Pan, Y. Gao, L. Chen, G. Gao, H. Dong et al., Targeting Autophagy Augments In Vitro and In Vivo Antimyeloma Activity of DNA-Damaging Chemotherapy, Clinical Cancer Research, vol.17, issue.10, pp.3248-3258, 2011.
DOI : 10.1158/1078-0432.CCR-10-0890

A. Puissant, G. Robert, and P. Auberger, Targeting autophagy to fight hematopoietic malignancies, Cell Cycle, vol.9, issue.17, pp.3470-3478, 2010.
DOI : 10.4161/cc.9.17.13048

A. Crawford, R. Riggins, A. Shajahan, A. Zwart, and C. R. , Co-Inhibition of BCL-W and BCL2 Restores Antiestrogen Sensitivity through BECN1 and Promotes an Autophagy-Associated Necrosis, PLoS ONE, vol.41, issue.1, p.8604, 2010.
DOI : 10.1371/journal.pone.0008604.s005

J. Moreaux, B. Klein, R. Bataille, G. Descamps, S. Maïga et al., A high-risk signature for patients with multiple myeloma established from the molecular classification of human myeloma cell lines, Haematologica, vol.96, issue.4, pp.574-582, 2011.
DOI : 10.3324/haematol.2010.033456

URL : https://hal.archives-ouvertes.fr/inserm-00550232

R. Fonseca, P. Bergsagel, J. Drach, J. Shaughnessy, N. Gutierrez et al., International Myeloma Working Group molecular classification of multiple myeloma: spotlight review, Leukemia, vol.91, issue.12, pp.2210-2221, 2009.
DOI : 10.1016/j.beha.2007.08.004

H. Schmidlin, S. Diehl, and B. Blom, New insights into the regulation of human B-cell differentiation, Trends in Immunology, vol.30, issue.6, pp.277-285, 2009.
DOI : 10.1016/j.it.2009.03.008

S. Silvente-poirot and M. Poirot, Cholesterol epoxide hydrolase and cancer, Current Opinion in Pharmacology, vol.12, issue.6, pp.696-703, 2012.
DOI : 10.1016/j.coph.2012.07.007

URL : https://hal.archives-ouvertes.fr/inserm-00743463

T. Berrodin, Q. Shen, E. Quinet, M. Yudt, L. Freedman et al., Identification of 5??,6??-Epoxycholesterol as a Novel Modulator of Liver X Receptor Activity, Molecular Pharmacology, vol.78, issue.6, pp.1046-1058, 2010.
DOI : 10.1124/mol.110.065193

L. Vedin, S. Lewandowski, P. Parini, J. Gustafsson, and K. Steffensen, The oxysterol receptor LXR inhibits proliferation of human breast cancer cells, Carcinogenesis, vol.30, issue.4, pp.575-579, 2009.
DOI : 10.1093/carcin/bgp029

N. Gutiérrez, E. Ocio, J. De-las-rivas, P. Maiso, M. Delgado et al., Gene expression profiling of B lymphocytes and plasma cells from Waldenstr??m's macroglobulinemia: comparison with expression patterns of the same cell counterparts from chronic lymphocytic leukemia, multiple myeloma and normal individuals, Leukemia, vol.21, issue.3, pp.541-549, 2007.
DOI : 10.1038/sj.leu.2404520

J. Storch and Z. Xu, Niemann???Pick C2 (NPC2) and intracellular cholesterol trafficking, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1791, issue.7, pp.671-678, 2009.
DOI : 10.1016/j.bbalip.2009.02.001

K. Peake and J. Vance, Defective cholesterol trafficking in Niemann-Pick C-deficient cells, FEBS Letters, vol.51, issue.13, pp.2731-2739, 2010.
DOI : 10.1016/j.febslet.2010.04.047

K. Liu and M. Czaja, Regulation of lipid stores and metabolism by lipophagy, Cell Death and Differentiation, vol.42, issue.1, pp.3-11, 2013.
DOI : 10.1371/journal.pone.0025269

M. Laplante and D. Sabatini, An Emerging Role of mTOR in Lipid Biosynthesis, Current Biology, vol.19, issue.22, pp.1046-1052, 2009.
DOI : 10.1016/j.cub.2009.09.058

S. Ricoult and B. Manning, The multifaceted role of mTORC1 in the control of lipid metabolism, EMBO reports, vol.106, issue.3, pp.242-251, 2013.
DOI : 10.1126/scisignal.267pe24

M. Poirot, P. De-medina, F. Delarue, J. Perie, A. Klaebe et al., Synthesis, binding and structure???affinity studies of new ligands for the microsomal anti-estrogen binding site (AEBS), Bioorganic & Medicinal Chemistry, vol.8, issue.8, pp.2007-2016, 2000.
DOI : 10.1016/S0968-0896(00)00119-X

URL : https://hal.archives-ouvertes.fr/inserm-00090778

A. Petiot, E. Ogier-denis, E. Blommaart, A. Meijer, and P. Codogno, Distinct Classes of Phosphatidylinositol 3'-Kinases Are Involved in Signaling Pathways That Control Macroautophagy in HT-29 Cells, Journal of Biological Chemistry, vol.275, issue.2, pp.992-998, 2000.
DOI : 10.1074/jbc.275.2.992

B. Payré, P. De-medina, N. Boubekeur, L. Mhamdi, J. Bertrand-michel et al., Microsomal antiestrogen-binding site ligands induce growth control and differentiation of human breast cancer cells through the modulation of cholesterol metabolism, Molecular Cancer Therapeutics, vol.7, issue.12, pp.3707-3718, 2008.
DOI : 10.1158/1535-7163.MCT-08-0507