P. Ascenzi, A. Bocedi, and M. Marino, Structure???function relationship of estrogen receptor ?? and ??: Impact on human health, Molecular Aspects of Medicine, vol.27, issue.4, pp.299-402, 2006.
DOI : 10.1016/j.mam.2006.07.001

S. Ali, R. Coombes, L. Helguero, M. Faulds, J. Gustafsson et al., Estrogen receptor alpha in human breast cancer: occurrence and significance Estrogen receptors alfa (ERalpha) and beta (ERbeta) differentially regulate proliferation and apoptosis of the normal murine mammary epithelial cell line HC11 PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients, J Mammary Gland Biol Neoplasia Oncogene Cancer Cell Nat Rev Cancer, vol.52465, pp.271-816605, 2000.

L. Romancer, M. Poulard, C. Cohen, P. Sentis, S. Renoir et al., Cracking the Estrogen Receptor's Posttranslational Code in Breast Tumors, Endocrine Reviews, vol.32, issue.5, pp.597-622, 2011.
DOI : 10.1210/er.2010-0016

W. Pratt, D. Toft, N. Heldring, A. Pike, S. Andersson et al., Estrogen receptors: how do they signal and what are their targets Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter Nuclear receptor coregulators: judges, juries, and executioners of cellular regulation The transcriptional coactivators p300 and CBP are histone acetyltransferases, Endocr Rev Physiol Rev Cell Mol Cell Cell, vol.18871151027118712, pp.306-60905, 1996.

X. Yang, V. Ogryzko, J. Nishikawa, B. Howard, and Y. Nakatani, A p300/CBP-associated factor that competes with the adenoviral oncoprotein E1A [13] O'Malley BW. Coregulators: from whence came these "master genes, Nature Mol Endocrinol, vol.3822114, pp.319-241009, 1996.

E. Musgrove, C. Caldon, J. Barraclough, A. Stone, R. Sutherland et al., Cyclin D as a therapeutic target in cancer Estrogen receptor beta inhibits human breast cancer cell proliferation and tumor formation by causing a G2 cell cycle arrest Estrogen receptor beta inhibits 17beta-estradiol-stimulated proliferation of the breast cancer cell line T47D Estrogen receptor alpha and beta subtype expression and transactivation capacity are differentially affected by receptor-, hsp90-and immunophilin-ligands in human breast cancer cells, Nat Rev Cancer Cancer Res Proc Natl Acad Sci U S A J Steroid Biochem Mol Biol, vol.1115641610194, pp.558-72423, 2004.

O. Grober, M. Mutarelli, G. Giurato, M. Ravo, L. Cicatiello et al., Global analysis of estrogen receptor beta binding to breast cancer cell genome reveals an extensive interplay with estrogen receptor alpha for target gene regulation, BMC Genomics, vol.37, issue.Database issue, p.36, 2011.
DOI : 10.1093/nar/gkn875

J. Matthews, J. Gustafsson, C. Bouclier, V. Marsaud, O. Bawa et al., Estrogen signaling: a subtle balance between ER alpha and ER beta Coadministration of nanosystems of short silencing RNAs targeting oestrogen receptor alpha and anti-oestrogen synergistically induces tumour growth inhibition in human breast cancer xenografts, Mol Interv Breast Cancer Res Treat, vol.3122, pp.281-92145, 2003.

C. Pequeux, I. Raymond-letron, S. Blacher, F. Boudou, M. Adlanmerini et al., Stromal Estrogen Receptor-alpha Promotes Tumor Growth by Normalizing an Increased Angiogenesis, Cancer Res, vol.7222, pp.3010-3019, 2012.
DOI : 10.1158/0008-5472.can-11-3768

URL : http://orbi.ulg.ac.be/jspui/handle/2268/124721

E. Levin and R. Pietras, Estrogen receptors outside the nucleus in breast cancer, Breast Cancer Research and Treatment, vol.9, issue.3, pp.351-61, 2008.
DOI : 10.1007/s10549-007-9618-4

T. Simoncini, A. Hafezi-moghadam, D. Brazil, K. Ley, W. Chin et al., Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase Caveolin-1, mammary stem cells, and estrogen-dependent breast cancers Estrogen receptor-dependent activation of AP-1 via non-genomic signalling, Nature Cancer Res Nucl Recept, vol.4072466252, pp.538-4110647, 2000.

M. Marino, F. Acconcia, F. Bresciani, A. Weisz, and A. Trentalance, Distinct Nongenomic Signal Transduction Pathways Controlled by 17beta -Estradiol Regulate DNA Synthesis and Cyclin D1 Gene Transcription in HepG2 Cells, Molecular Biology of the Cell, vol.13, issue.10, pp.3720-3729, 2002.
DOI : 10.1091/mbc.E02-03-0153

L. Kang, X. Zhang, Y. Xie, Y. Tu, D. Wang et al., Involvement of estrogen receptor variant ER-alpha36, not GPR30, in nongenomic estrogen signaling Minireview: G Protein-Coupled Estrogen Receptor-1, GPER-1: Its Mechanism of Action and Role in Female Reproductive Cancer, Renal and Vascular Physiology, Mol Endocrinol Endocrinology, vol.242815329, pp.709-212953, 2010.

E. Filardo, J. Quinn, A. Frackelton, J. Bland, K. Dennis et al., Estrogen action via the G protein-coupled receptor, GPR30: stimulation of adenylyl cyclase and cAMP-mediated attenuation of the epidermal growth factor receptor-to-MAPK signaling axis Identification of a GPER/GPR30 antagonist with improved estrogen receptor counterselectivity, MIBE acts as antagonist ligand of both estrogen receptor alpha and GPER in breast cancer cells, pp.70-84358, 2002.

V. S. Jordan, D. Lonard, Z. Nawaz, C. Smith, O. Malley et al., The 26S proteasome is required for estrogen receptor-alpha and coactivator turnover and for efficient estrogen receptor-alpha transactivation Various phosphorylation pathways, depending on agonist and antagonist binding to endogenous estrogen receptor alpha (ERalpha), differentially affect ERalpha extractability, proteasome-mediated stability, and transcriptional activity in human breast cancer cells, J Natl Cancer Inst Mol Cell Mol Endocrinol, vol.9951735, pp.350-6939, 2000.

S. Ali and R. Coombes, ENDOCRINE-RESPONSIVE BREAST CANCER AND STRATEGIES FOR COMBATING RESISTANCE, Nature Reviews Cancer, vol.104, issue.2, pp.101-113, 2002.
DOI : 10.1126/science.277.5331.1508

R. Clarke, M. Liu, K. Bouker, Z. Gu, R. Lee et al., Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling, Oncogene, vol.22, issue.47, pp.7316-7355, 2003.
DOI : 10.1038/sj.onc.1206937

V. Jordan, O. Malley, B. Nicholson, R. Hutcheson, I. Hiscox et al., Selective estrogen-receptor modulators and antihormonal resistance in breast cancer Growth factor signalling and resistance to selective oestrogen receptor modulators and pure antioestrogens: the use of anti-growth factor therapies to treat or delay endocrine resistance in breast cancer Growth factor signalling in endocrine and anti-growth factor resistant breast cancer, J Clin Oncol Endocr Relat Cancer Rev Endocr Metab Disord, vol.2512398, issue.1, pp.5815-5839, 2005.

E. Musgrove, R. Sutherland, R. Sutherland, L. Murphy, S. Foo et al., Highaffinity anti-oestrogen binding site distinct from the oestrogen receptor Molecular characterization of the microsomal tamoxifen binding site, Ligands of the antiestrogen-binding site induce active cell death and autophagy in human breast cancer cells through the modulation of cholesterol metabolism, pp.631-43273, 1980.

L. Reyno, L. Seymour, D. Tu, S. Dent, K. Gelmon et al., -Diethyl-2-[4-(Phenylmethyl) Phenoxy]Ethanamine (BMS-217380-01) Combined With Doxorubicin Versus Doxorubicin Alone in Metastatic/Recurrent Breast Cancer: National Cancer Institute of Canada Clinical Trials Group Study MA.19, Journal of Clinical Oncology, vol.22, issue.2, pp.269-76, 2004.
DOI : 10.1200/JCO.2003.04.075

J. Liu, D. Tu, J. Dancey, L. Reyno, K. Pritchard et al., Quality of life analyses in a clinical trial of DPPE (tesmilifene) plus doxorubicin versus doxorubicin in patients with advanced or metastatic breast cancer: NCIC CTG Trial MA.19 [46] (EBCTCG) Ebctcg. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials, Breast Cancer Res Treat Lancet, vol.10036547, pp.263-711687, 2005.

E. Henson and S. Gibson, Surviving cell death through epidermal growth factor (EGF) signal transduction pathways: Implications for cancer therapy, Cellular Signalling, vol.18, issue.12, pp.2089-97, 2006.
DOI : 10.1016/j.cellsig.2006.05.015

Y. Wang, D. Lonard, Y. Yu, D. Chow, T. Palzkill et al., Small Molecule Inhibition of the Steroid Receptor Coactivators, SRC-3 and SRC-1, Molecular Endocrinology, vol.25, issue.12, pp.2041-53, 2011.
DOI : 10.1210/me.2011-1222

P. Goodwin, B. Manning, L. Cantley, I. Hutcheson, L. Goddard et al., Insulin in the adjuvant breast cancer setting: a novel therapeutic target for lifestyle and pharmacologic interventions AKT/PKB signaling: navigating downstream Fulvestrant-induced expression of ErbB3 and ErbB4 receptors sensitizes oestrogen receptorpositive breast cancer cells to heregulin beta1, J Clin Oncol Cell Breast Cancer Res, vol.2612913, pp.833-41261, 2007.

R. Sutherland, P. Van-de-velde, F. Nique, J. Bremaud, M. Hameau et al., Endocrine resistance in breast cancer: new roles for ErbB3 and ErbB4 Exploration of the therapeutic potential of the antiestrogen RU 58668 in breast cancer treatment, Breast Cancer Res Ann N Y Acad Sci, vol.1376154, pp.106-53, 1995.

J. Menendez, I. Mehmi, R. Lupu, V. Kaklamani, K. Siziopikou et al., Trastuzumab in combination with heregulin-activated Her-2 (erbB-2) triggers a receptor-enhanced chemosensitivity effect in the absence of Her-2 overexpression Pilot neoadjuvant trial in HER2 positive breast cancer with combination of nab-paclitaxel and lapatinib The nuclear receptor coactivator amplified in breast cancer-1 is required for Neu (ErbB2/HER2) activation, signaling, and mammary tumorigenesis in mice, J Clin Oncol Breast Cancer Res Treat Cancer Res, vol.24132566857, pp.3735-46833, 2006.

T. Lahusen, R. Henke, B. Kagan, A. Wellstein, and A. Riegel, The role and regulation of the nuclear receptor co-activator AIB1 in breast cancer, Breast Cancer Research and Treatment, vol.29, issue.2, pp.225-262, 2009.
DOI : 10.1007/s10549-009-0405-2

G. Urbinati, V. Marsaud, V. Plassat, E. Fattal, S. Lesieur et al., Liposomes loaded with histone deacetylase inhibitors for breast cancer therapy, International Journal of Pharmaceutics, vol.397, issue.1-2, pp.184-93, 2010.
DOI : 10.1016/j.ijpharm.2010.06.046

V. Duong, A. Licznar, R. Margueron, N. Boulle, M. Busson et al., ERalpha and ERbeta expression and transcriptional activity are differentially regulated by HDAC inhibitors, Oncogene, vol.2560, pp.1799-806, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00143967

Q. Zhou, P. Atadja, and N. Davidson, Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation, Cancer Biology & Therapy, vol.6, issue.1, pp.64-961, 2007.
DOI : 10.4161/cbt.6.1.3549

E. Jang, S. Lim, E. Lee, G. Jeong, T. Kim et al., The histone deacetylase inhibitor trichostatin A sensitizes estrogen receptor alpha-negative breast cancer cells to tamoxifen, Oncogene, vol.2362, pp.1724-1760, 2004.

S. Minucci, P. Pelicci, A. Licznar, G. Lazennec, F. Vignon et al., Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer Oestrogen receptor alpha increases p21(WAF1/CIP1) gene expression and the antiproliferative activity of histone deacetylase inhibitors in human breast cancer cells, Nat Rev Cancer J Endocrinol, vol.617964, pp.38-5141, 2003.

V. Duong, C. Bret, L. Altucci, A. Mai, C. Duraffourd et al., Specific activity of class II histone deacetylases in human breast cancer cells A novel GRK2/HDAC6 interaction modulates cell spreading and motility Regulation of ERBB2 by oestrogen receptor-PAX2 determines response to tamoxifen FOXA1 is a key determinant of estrogen receptor function and endocrine response Differential oestrogen receptor binding is associated with clinical outcome in breast cancer, Mol Cancer Res Embo J Nature Nat Genet Nature Breast Cancer Res Treat, vol.6314566743684816913270, pp.1908-19856, 2008.

L. Romancer, M. Treilleux, I. Leconte, N. Robin-lespinasse, Y. Sentis et al., Regulation of Estrogen Rapid Signaling through Arginine Methylation by PRMT1, Molecular Cell, vol.31, issue.2, pp.212-233, 2008.
DOI : 10.1016/j.molcel.2008.05.025

Y. Huang, S. Vasilatos, L. Boric, P. Shaw, and N. Davidson, Inhibitors of histone demethylation and histone deacetylation cooperate in regulating gene expression and inhibiting growth in human breast cancer cells, Breast Cancer Research and Treatment, vol.225, issue.3, pp.777-89, 2012.
DOI : 10.1007/s10549-011-1480-8

Y. Huang, E. Greene, M. Stewart, T. Goodwin, A. Baylin et al., Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes Lysine-specific demethylase 1 (LSD1/KDM1A/AOF2/BHC110) is expressed and is an epigenetic drug target in chondrosarcoma, Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma, Proc Natl Acad Sci U S A Hum Pathol, vol.104734374, pp.8023-81300, 2007.

R. Vadlamudi, R. Kumar, P. Mnar-brann, D. Zhang, Q. Wang et al., PELP1--a novel estrogen receptor-interacting protein PELP1 is a reader of histone H3 methylation that facilitates oestrogen receptor-alpha target gene activation by regulating lysine demethylase 1 specificity The prognostic significance of PELP1 expression in invasive breast cancer with emphasis on the ER-positive luminal-like subtype, Nucl Recept Signal Mol Cell Endocrinol EMBO Rep Breast Cancer Res Treat, vol.52901177120, pp.2-7438, 2007.

K. Subbaramaiah, L. Howe, E. Port, E. Brogi, J. Fishman et al., HER-2/neu status is a determinant of mammary aromatase activity in vivo: evidence for a cyclooxygenase-2- dependent mechanism Targeting the PELP1-KDM1 axis as a potential therapeutic strategy for breast cancer Bennani-Baiti IM. Integration of ERalpha-PELP1-HER2 signaling by LSD1 (KDM1A/AOF2) offers combinatorial therapeutic opportunities to circumventing hormone resistance in breast cancer LKB1 is recruited to the p21/WAF1 promoter by p53 to mediate transcriptional activation, ZM. Enhanced expression of LKB1 in breast cancer cells attenuates angiogenesis, invasion, and metastatic potential, pp.5504-1111210701, 2006.

D. Hardie, AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy, Nature Reviews Molecular Cell Biology, vol.367, issue.10, pp.774-85, 2007.
DOI : 10.1038/nrm2249

Z. Shen, X. Wen, F. Lan, Z. Shen, Z. Shao et al., The tumor suppressor gene LKB1 is associated with prognosis in human breast carcinoma LKB1 catalytic activity contributes to estrogen receptor alpha signaling Liver kinase B1 expression (LKB1) is repressed by estrogen receptor alpha (ERalpha) in MCF-7 human breast cancer cells Metformin inhibits aromatase expression in human breast adipose stromal cells via stimulation of AMP-activated protein kinase LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis and inhibition of migration and invasion of breast cancer cells, Clin Cancer Res Mol Biol Cell Biochem Biophys Res Commun Breast Cancer Res Treat Oncogene, vol.885208641787123882889, pp.2085-902785, 2002.

X. Wu, H. Chen, B. Parker, E. Rubin, T. Zhu et al., HOXB7, a Homeodomain Protein, Is Overexpressed in Breast Cancer and Confers Epithelial-Mesenchymal Transition, Cancer Research, vol.66, issue.19, pp.9527-9561, 2006.
DOI : 10.1158/0008-5472.CAN-05-4470

K. Jin, X. Kong, T. Shah, M. Penet, F. Wildes et al., The HOXB7 protein renders breast cancer cells resistant to tamoxifen through activation of the EGFR pathway Transducinlike enhancer protein 1 mediates estrogen receptor binding and transcriptional activity in breast cancer cells, Proc Natl Acad Sci U S A Proc Natl Acad Sci U S A, vol.10910992, pp.2736-412748, 2012.

J. Couse, K. Korach, G. Lazennec, D. Bresson, A. Lucas et al., Estrogen receptor null mice: what have we learned and where will they lead us? ER beta inhibits proliferation and invasion of breast cancer cells, Endocr Rev Endocrinology, vol.2014294, pp.358-4174120, 1999.

J. Hartman, K. Lindberg, A. Morani, J. Inzunza, A. Strom et al., Estrogen receptor beta inhibits angiogenesis and growth of T47D breast cancer xenografts, Cancer Res, vol.6695, pp.11207-11220, 2006.

G. Lazennec, L. Hodges-gallagher, C. Valentine, S. Bader, and P. Kushner, Estrogen receptor beta, a possible tumor suppressor involved in ovarian carcinogenesis Estrogen receptor beta increases the efficacy of antiestrogens by effects on apoptosis and cell cycling in breast cancer cells, Cancer Lett Breast Cancer Res Treat, vol.23196109, pp.151-7241, 2006.

K. Lindberg, L. Helguero, Y. Omoto, J. Gustafsson, L. Haldosen et al., Estrogen receptor beta represses Akt signaling in breast cancer cells via downregulation of HER2/HER3 and upregulation of PTEN: implications for tamoxifen sensitivity Mechanisms of growth arrest by c-myc antisense oligonucleotides in MCF-7 breast cancer cells: implications for the antiproliferative effects of antiestrogens, Breast Cancer Res Cancer Res, vol.136299, pp.3126-3157, 2002.

G. Castoria, A. Migliaccio, A. Bilancio, D. Domenico, M. De-falco et al., PI3-kinase in concert with Src promotes the S-phase entry of oestradiol-stimulated MCF-7 cells, The EMBO Journal, vol.20, issue.21, pp.6050-6059, 2001.
DOI : 10.1093/emboj/20.21.6050

S. Hiscox, L. Morgan, T. Green, D. Barrow, J. Gee et al., Elevated Src activity promotes cellular invasion and motility in tamoxifen resistant breast cancer cells, Breast Cancer Research and Treatment, vol.278, issue.3, pp.263-74, 2006.
DOI : 10.1007/s10549-005-9120-9

X. Zhang, Q. Wang, W. Gerald, C. Hudis, L. Norton et al., Latent Bone Metastasis in Breast Cancer Tied to Src-Dependent Survival Signals, Cancer Cell, vol.16, issue.1, pp.67-78, 2009.
DOI : 10.1016/j.ccr.2009.05.017

M. Herynk, A. Beyer, Y. Cui, H. Weiss, E. Anderson et al., Cooperative action of tamoxifen and c-Src inhibition in preventing the growth of estrogen receptor-positive human breast cancer cells, Molecular Cancer Therapeutics, vol.5, issue.12, pp.3023-3054, 2006.
DOI : 10.1158/1535-7163.MCT-06-0394

S. Hiscox, L. Morgan, T. Green, and R. Nicholson, Src as a therapeutic target in anti-hormone/anti-growth factor-resistant breast cancer, Endocrine Related Cancer, vol.13, issue.Supplement 1, pp.53-62, 2006.
DOI : 10.1677/erc.1.01297

S. Wander, B. Hennessy, and J. Slingerland, Next-generation mTOR inhibitors in clinical oncology: how pathway complexity informs therapeutic strategy, Journal of Clinical Investigation, vol.121, issue.4, pp.1231-1272, 2011.
DOI : 10.1172/JCI44145

S. Zhang, W. Huang, P. Li, H. Guo, S. Poh et al., Abstract LB-379: Combating trastuzumab resistance by targeting Src, a common node downstream of multiple resistance pathways, Cancer Research, vol.71, issue.8 Supplement, pp.461-470, 2011.
DOI : 10.1158/1538-7445.AM2011-LB-379

G. Lazennec and R. A. , Chemokines and chemokine receptors: new insights into cancer-related inflammation, Trends in Molecular Medicine, vol.16, issue.3, pp.133-177, 2010.
DOI : 10.1016/j.molmed.2010.01.003

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2840699

I. Bieche, C. Chavey, C. Andrieu, M. Busson, S. Vacher et al., CXC chemokines located in the 4q21 region are up-regulated in breast cancer, Endocrine Related Cancer, vol.14, issue.4, pp.1039-52, 2007.
DOI : 10.1677/erc.1.01301

URL : https://hal.archives-ouvertes.fr/inserm-00153602

C. Chavey, F. Bibeau, S. Gourgou-bourgade, S. Burlinchon, F. Boissiere et al., Oestrogen receptor negative breast cancers exhibit high cytokine content, Breast Cancer Research, vol.9, issue.1, p.15, 2007.
DOI : 10.1186/bcr1648

URL : https://hal.archives-ouvertes.fr/inserm-00143810

C. Chavey, M. Muhlbauer, C. Bossard, A. Freund, S. Durand et al., Interleukin-8 Expression Is Regulated by Histone Deacetylases through the Nuclear Factor-??B Pathway in Breast Cancer, Molecular Pharmacology, vol.74, issue.5, pp.1359-66, 2008.
DOI : 10.1124/mol.108.047332

A. Boudot, G. Kerdivel, D. Habauzit, J. Eeckhoute, L. Dily et al., Differential Estrogen-Regulation of CXCL12 Chemokine Receptors, CXCR4 and CXCR7, Contributes to the Growth Effect of Estrogens in Breast Cancer Cells, PLoS ONE, vol.132, issue.6, p.20898, 2011.
DOI : 10.1371/journal.pone.0020898.s002

URL : https://hal.archives-ouvertes.fr/hal-00605459

T. Kobayashi, H. Tsuda, T. Moriya, T. Yamasaki, R. Kikuchi et al., Expression pattern of stromal cell-derived factor-1 chemokine in invasive breast cancer is correlated with estrogen receptor status and patient prognosis, Breast Cancer Research and Treatment, vol.3, issue.12, pp.733-778, 2010.
DOI : 10.1007/s10549-009-0672-y

S. Sengupta, R. Schiff, and B. Katzenellenbogen, Post-transcriptional regulation of chemokine receptor CXCR4 by estrogen in HER2 overexpressing, estrogen receptor-positive breast cancer cells, Breast Cancer Research and Treatment, vol.66, issue.2, pp.243-51, 2009.
DOI : 10.1007/s10549-008-0186-z

L. Rhodes, S. Short, N. Neel, V. Salvo, Y. Zhu et al., Cytokine Receptor CXCR4 Mediates Estrogen-Independent Tumorigenesis, Metastasis, and Resistance to Endocrine Therapy in Human Breast Cancer, Cancer Research, vol.71, issue.2, pp.603-616, 2011.
DOI : 10.1158/0008-5472.CAN-10-3185

K. Sauve, J. Lepage, M. Sanchez, N. Heveker, and A. Tremblay, Positive Feedback Activation of Estrogen Receptors by the CXCL12-CXCR4 Pathway, Cancer Research, vol.69, issue.14, pp.5793-800, 2009.
DOI : 10.1158/0008-5472.CAN-08-4924

M. Pollak, Insulin and insulin-like growth factor signalling in neoplasia, Nature Reviews Cancer, vol.5, issue.12, pp.915-943, 2008.
DOI : 10.2174/156800907781386623

J. Jones and D. Clemmons, Insulin-like growth factors and their binding proteins: biological actions, Endocr Rev, vol.16, pp.3-34, 1995.

J. Law, G. Habibi, K. Hu, H. Masoudi, M. Wang et al., Phosphorylated Insulin-Like Growth Factor-I/Insulin Receptor Is Present in All Breast Cancer Subtypes and Is Related to Poor Survival, Cancer Research, vol.68, issue.24, pp.10238-10284, 2008.
DOI : 10.1158/0008-5472.CAN-08-2755

J. Lisztwan, A. Pornon, B. Chen, S. Chen, and D. Evans, The aromatase inhibitor letrozole and inhibitors of insulin-like growth factor I receptor synergistically induce apoptosis in in vitro models of estrogen-dependent breast cancer, Breast Cancer Research, vol.102, issue.suppl 1, p.56, 2008.
DOI : 10.1007/s10549-006-9307-8

J. Gao, Y. Chang, B. Jallal, and J. Viner, Targeting the Insulin-like Growth Factor Axis for the Development of Novel Therapeutics in Oncology, Cancer Research, vol.72, issue.1, pp.3-12, 2012.
DOI : 10.1158/0008-5472.CAN-11-0550

A. Casa, A. Potter, S. Malik, Z. Lazard, I. Kuiatse et al., Estrogen and insulin-like growth factor-I (IGF-I) independently down-regulate critical repressors of breast cancer growth, Breast Cancer Research and Treatment, vol.108, issue.1, pp.61-73, 2012.
DOI : 10.1007/s10549-011-1540-0

J. Renoir, C. Radanyi, L. Faber, and E. Baulieu, The non-DNA-binding heterooligomeric form of mammalian steroid hormone receptors contains a hsp90-bound 59-kilodalton protein, J Biol Chem, vol.265, pp.10740-10745, 1990.

B. Freeman, S. Felts, D. Toft, and K. Yamamoto, The p23 molecular chaperones act at a late step in intracellular receptor action to differentially affect ligand efficacies, Genes Dev, vol.14, pp.422-456, 2000.

L. Neckers, Heat shock protein 90: The cancer chaperone, Journal of Biosciences, vol.23, issue.3, pp.517-547, 2007.
DOI : 10.1007/s12038-007-0051-y

B. Blagg and T. Kerr, Hsp90 inhibitors: Small molecules that transform the Hsp90 protein folding machinery into a catalyst for protein degradation, Medicinal Research Reviews, vol.100, issue.3, pp.310-348, 2006.
DOI : 10.1002/med.20052

C. Radanyi, L. Bras, G. Marsaud, V. Peyrat, J. Messaoudi et al., Antiproliferative and apoptotic activities of tosylcyclonovobiocic acids as potent heat shock protein 90 inhibitors in human cancer cells, Cancer Letters, vol.274, issue.1, pp.88-94, 2009.
DOI : 10.1016/j.canlet.2008.09.001

D. Audisio, S. Messaoudi, L. Cegielkowski, J. Peyrat, J. Brion et al., Discovery and Biological Activity of 6BrCaQ as an Inhibitor of the Hsp90 Protein Folding Machinery, ChemMedChem, vol.306, issue.5, pp.804-819, 2011.
DOI : 10.1002/cmdc.201000489

X. Yu, X. Zhang, I. Dhakal, M. Beggs, S. Kadlubar et al., Induction of cell proliferation and survival genes by estradiol-repressed microRNAs in breast cancer cells, BMC Cancer, vol.304, issue.1, p.29, 2012.
DOI : 10.1016/j.canlet.2011.02.003

G. Urbinati, V. Marsaud, and J. Renoir, Anticancer Drugs in Liposomal Nanodevices: A Target Delivery for a Targeted Therapy, Current Topics in Medicinal Chemistry, vol.12, issue.15, 2012.
DOI : 10.2174/156802612803531423

D. Drummond, C. Marx, Z. Guo, G. Scott, C. Noble et al., Enhanced Pharmacodynamic and Antitumor Properties of a Histone Deacetylase Inhibitor Encapsulated in Liposomes or ErbB2-Targeted Immunoliposomes, Clinical Cancer Research, vol.11, issue.9, pp.3392-401, 2005.
DOI : 10.1158/1078-0432.CCR-04-2445

I. Krop, P. Lorusso, K. Miller, S. Modi, Y. D. Rodriguez et al., A Phase II Study of Trastuzumab Emtansine in Patients With Human Epidermal Growth Factor Receptor 2???Positive Metastatic Breast Cancer Who Were Previously Treated With Trastuzumab, Lapatinib, an Anthracycline, a Taxane, and Capecitabine, Journal of Clinical Oncology, vol.30, issue.26, pp.3234-3275, 2012.
DOI : 10.1200/JCO.2011.40.5902

P. Munster, K. Thurn, S. Thomas, P. Raha, M. Lacevic et al., A phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer, British Journal of Cancer, vol.6, issue.12, pp.1828-1863, 2011.
DOI : 10.1038/bjc.2011.156

M. Anbalagan, L. Carrier, S. Glodowski, D. Hangauer, B. Shan et al., KX-01, a novel Src kinase inhibitor directed toward the peptide substrate site, synergizes with tamoxifen in estrogen receptor ?? positive breast cancer, Breast Cancer Research and Treatment, vol.93, issue.9, pp.391-409, 2012.
DOI : 10.1007/s10549-011-1513-3

F. Cai, C. Kohler, B. Zhang, M. Wang, W. Chen et al., Epigenetic Therapy for Breast Cancer, International Journal of Molecular Sciences, vol.12, issue.12, pp.4465-87, 2011.
DOI : 10.3390/ijms12074465

Q. Zhou, P. Shaw, and N. Davidson, Epigenetics meets estrogen receptor: regulation of estrogen receptor by direct lysine methylation, Endocrine Related Cancer, vol.16, issue.2, pp.319-342, 2009.
DOI : 10.1677/ERC-08-0305

K. Paech, P. Webb, G. Kuiper, S. Nilsson, J. Gustafsson et al., Differential Ligand Activation of Estrogen Receptors ER and ER at AP1 Sites, Science, vol.277, issue.5331, pp.1508-1518, 1997.
DOI : 10.1126/science.277.5331.1508

T. Peterson, S. Karmakar, M. Pace, T. Gao, and C. Smith, The Silencing Mediator of Retinoic Acid and Thyroid Hormone Receptor (SMRT) Corepressor Is Required for Full Estrogen Receptor ?? Transcriptional Activity, Molecular and Cellular Biology, vol.27, issue.17, pp.5933-5981, 2007.
DOI : 10.1128/MCB.00237-07