D. J. Slamon, G. M. Clark, S. G. Wong, W. J. Levin, A. Ullrich et al., Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene, Science, vol.235, issue.4785, pp.177-82, 1987.

E. M. Bublil and Y. Yarden, The EGF receptor family: spearheading a merger of signaling and therapeutics, Curr Opin Cell Biol, vol.19, issue.2, pp.124-158, 2007.

A. Citri and Y. Yarden, EGF-ERBB signalling: towards the systems level, Nat Rev Mol Cell Biol, vol.7, issue.7, pp.505-521, 2006.

Y. Yarden and M. X. Sliwkowski, Untangling the ErbB signalling network, Nat Rev Mol Cell Biol, vol.2, issue.2, pp.127-164, 2001.
DOI : 10.1038/35052073

, Haute Autorité de Santé-HERCEPTIN (trastuzumab), monoclonal antibody, 2017.

B. Cetin, M. Benekli, F. Dane, C. Boruban, M. Gumus et al., Lapatinib plus Capecitabine for HER2-Positive Advanced-Stage Breast Cancer in Elderly Women: Review of the Anatolian Society of Medical Oncology (ASMO) Experience. Breast Care Basel Switz, vol.8, pp.67-70, 2013.

E. A. Perez, E. H. Romond, V. J. Suman, J. Jeong, G. Sledge et al., Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831, J Clin Oncol Off J Am Soc Clin Oncol, vol.32, issue.33, pp.3744-52, 2014.

M. Ewertz, M. Jensen, K. Á. Gunnarsdóttir, I. Højris, E. H. Jakobsen et al., Effect of obesity on prognosis after early-stage breast cancer, J Clin Oncol Off J Am Soc Clin Oncol, vol.29, issue.1, pp.25-31, 2011.

B. Dave, I. Migliaccio, M. C. Gutierrez, M. Wu, G. C. Chamness et al., Loss of phosphatase and tensin homolog or phosphoinositol-3 kinase activation and response to trastuzumab or lapatinib in human epidermal growth factor receptor 2-overexpressing locally advanced breast cancers, J Clin Oncol Off J Am Soc Clin Oncol, vol.29, issue.2, pp.166-73, 2011.

B. Lauby-secretan, C. Scoccianti, D. Loomis, Y. Grosse, F. Bianchini et al., Body Fatness and Cancer-Viewpoint of the IARC Working Group, N Engl J Med, vol.375, issue.8, pp.794-802, 2016.

M. Arnold, M. Touillaud, L. Dossus, H. Freisling, F. Bray et al., Cancers in France in 2015 attributable to high body mass index, Cancer Epidemiol, vol.52, pp.15-24, 2018.

E. E. Calle, C. Rodriguez, K. Walker-thurmond, and M. J. Thun, Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults, N Engl J Med, vol.348, issue.17, pp.1625-1663, 2003.

K. H. Schmitz, M. L. Neuhouser, T. Agurs-collins, K. A. Zanetti, L. Cadmus-bertram et al., Impact of obesity on cancer survivorship and the potential relevance of race and ethnicity, J Natl Cancer Inst, vol.105, issue.18, pp.1344-54, 2013.

J. A. Crozier, A. Moreno-aspitia, K. V. Ballman, A. C. Dueck, B. A. Pockaj et al., Effect of body mass index on tumor characteristics and disease-free survival in patients from the HER2-positive adjuvant trastuzumab trial N9831. Cancer, vol.119, pp.2447-54, 2013.

M. N. Duong, A. Cleret, E. Matera, K. Chettab, D. Mathé et al., Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity, Breast Cancer Res BCR, vol.17, issue.1, p.57, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01820280

L. Formisano, L. Nappi, R. Rosa, R. Marciano, D. 'amato et al., Epidermal growth factor-receptor activation modulates Src-dependent resistance to lapatinib in breast cancer models, Breast Cancer Res BCR, vol.16, issue.3, p.45, 2014.
DOI : 10.1186/bcr3650

URL : https://breast-cancer-research.biomedcentral.com/track/pdf/10.1186/bcr3650

L. Liu, J. Greger, H. Shi, Y. Liu, J. Greshock et al., Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: activation of AXL, Cancer Res, vol.69, issue.17, pp.6871-6879, 2009.

D. L. Wheeler, S. Huang, T. J. Kruser, M. M. Nechrebecki, E. A. Armstrong et al., Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members, Oncogene, vol.27, issue.28, pp.3944-56, 2008.

W. Yu, D. Cao, Q. Li, H. Mei, Y. Hu et al., Adipocytes secreted leptin is a pro-tumor factor for survival of multiple myeloma under chemotherapy, Oncotarget, vol.7, issue.52, pp.86075-86, 2016.

J. W. Behan, J. P. Yun, M. P. Proektor, E. A. Ehsanipour, A. Arutyunyan et al., Adipocytes impair leukemia treatment in mice, Cancer Res, vol.69, pp.7867-74, 2009.

K. M. Nieman, H. A. Kenny, C. V. Penicka, A. Ladanyi, R. Buell-gutbrod et al., Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth, Nat Med, vol.17, issue.11, pp.1498-503, 2011.
DOI : 10.1038/nm.2492

URL : http://europepmc.org/articles/pmc4157349?pdf=render

M. Scaltriti, F. Rojo, A. Ocaña, J. Anido, M. Guzman et al., Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer, J Natl Cancer Inst, vol.99, issue.8, pp.628-666, 2007.

K. Tamura, C. Shimizu, T. Hojo, S. Akashi-tanaka, T. Kinoshita et al., Fc?R2A and 3A polymorphisms predict clinical outcome of trastuzumab in both neoadjuvant and metastatic settings in patients with HER2-positive breast cancer, Ann Oncol Off J Eur Soc Med Oncol, vol.22, issue.6, pp.1302-1309, 2011.

R. Nahta, T. Takahashi, N. T. Ueno, M. Hung, and F. J. Esteva, P27(kip1) down-regulation is associated with trastuzumab resistance in breast cancer cells, Cancer Res, vol.64, issue.11, pp.3981-3987, 2004.
DOI : 10.1158/0008-5472.can-03-3900

URL : http://cancerres.aacrjournals.org/content/64/11/3981.full.pdf

K. Berns, H. M. Horlings, B. T. Hennessy, M. Madiredjo, E. M. Hijmans et al., A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer, Cancer Cell, vol.12, issue.4, pp.395-402, 2007.

B. N. Rexer, A. Ham, C. Rinehart, S. Hill, M. Granja-ingram-n-de et al., Phosphoproteomic mass spectrometry profiling links Src family kinases to escape from HER2 tyrosine kinase inhibition, Oncogene, vol.30, issue.40, pp.4163-74, 2011.
DOI : 10.1038/onc.2011.130

URL : https://www.nature.com/articles/onc2011130.pdf

J. W. Polli, J. E. Humphreys, K. A. Harmon, S. Castellino, O. Mara et al., (methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine (GW572016, lapatinib) disposition and drug interactions, Drug Metab Dispos Biol Fate Chem, vol.36, issue.4, pp.695-701, 2008.
DOI : 10.1124/dmd.107.018374

W. Xia, S. Bacus, P. Hegde, I. Husain, J. Strum et al., A model of acquired autoresistance to a potent ErbB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in breast cancer, Proc Natl Acad Sci, vol.103, issue.20, pp.7795-800, 2006.

L. Tang, Y. Wang, A. Strom, J. Gustafsson, and X. Guan, Lapatinib induces p27(Kip1)-dependent G arrest through both transcriptional and post-translational mechanisms, Cell Cycle Georget Tex, vol.12, issue.16, pp.2665-74, 2013.
DOI : 10.4161/cc.25728

URL : http://europepmc.org/articles/pmc3865056?pdf=render

Z. Liu, J. Xu, J. He, H. Liu, P. Lin et al., Mature adipocytes in bone marrow protect myeloma cells against chemotherapy through autophagy activation, Oncotarget, vol.6, issue.33, p.34329, 2015.
DOI : 10.18632/oncotarget.6020

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=6020&path%5B%5D=14598

D. Angel, R. E. Blando, J. M. Hogan, M. G. Sandoval, M. A. Lansakara et al., Stearoyl gemcitabine nanoparticles overcome obesity-induced cancer cell resistance to gemcitabine in a mouse postmenopausal breast cancer model, Cancer Biol Ther, vol.14, issue.4, pp.357-64, 2013.

P. Malvi, B. Chaube, S. V. Singh, N. Mohammad, V. Pandey et al., Weight control interventions improve therapeutic efficacy of dacarbazine in melanoma by reversing obesity-induced drug resistance, Cancer Metab, vol.4, p.21, 2016.

N. Grabinski and F. Ewald, Ibrutinib (ImbruvicaTM) potently inhibits ErbB receptor phosphorylation and cell viability of ErbB2-positive breast cancer cells, Invest New Drugs, vol.32, issue.6, pp.1096-104, 2014.
DOI : 10.1007/s10637-014-0141-2

F. Solca, G. Dahl, A. Zoephel, G. Bader, M. Sanderson et al., Target binding properties and cellular activity of afatinib (BIBW 2992), an irreversible ErbB family blocker, J Pharmacol Exp Ther, vol.343, issue.2, pp.342-50, 2012.

P. R. Gavine, L. Mooney, E. Kilgour, A. P. Thomas, K. Al-kadhimi et al., AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family, Cancer Res, vol.72, issue.8, pp.2045-56, 2012.

S. M. Johansson, E. Lindgren, J. Yang, A. W. Herling, and B. B. Fredholm, Adenosine A1 receptors regulate lipolysis and lipogenesis in mouse adipose tissue-Interactions with insulin, Eur J Pharmacol, vol.597, issue.1, pp.92-101, 2008.

H. Kather, Pathways of purine metabolism in human adipocytes. Further evidence against a role of adenosine as an endogenous regulator of human fat cell function, J Biol Chem, vol.265, issue.1, pp.96-102, 1990.

H. I. Cheong, K. Asosingh, O. R. Stephens, K. A. Queisser, W. Xu et al., Hypoxia sensing through ?-adrenergic receptors, JCI Insight, vol.1, issue.21, 2017.
DOI : 10.1172/jci.insight.90240

URL : http://insight.jci.org/articles/view/90240/files/pdf

J. Tan, E. Buache, M. Chenard, N. Dali-youcef, and M. Rio, Adipocyte is a non-trivial, dynamic partner of breast cancer cells, Int J Dev Biol, vol.55, issue.7-9, pp.851-860, 2011.

S. W. Brady, J. Zhang, M. Tsai, and D. Yu, PI3K-independent mTOR activation promotes lapatinib resistance and IAP expression that can be effectively reversed by mTOR and Hsp90 inhibition, Cancer Biol Ther, vol.16, issue.3, pp.402-413, 2015.
DOI : 10.1080/15384047.2014.1002693

URL : https://www.tandfonline.com/doi/pdf/10.1080/15384047.2014.1002693?needAccess=true

A. Vazquez-martin, C. Oliveras-ferraros, R. Colomer, J. Brunet, and J. A. Menendez, Low-scale phosphoproteome analyses identify the mTOR effector p70 S6 kinase 1 as a specific biomarker of the dual-HER1/HER2 tyrosine kinase inhibitor lapatinib (Tykerb®) in human breast carcinoma cells, Ann Oncol, vol.19, issue.6, pp.1097-109, 2008.

Y. L. Wang-fisher, J. Han, and W. Guo, Acipimox stimulates leptin production from isolated rat adipocytes, J Endocrinol, vol.174, issue.2, pp.267-72, 2002.
DOI : 10.1677/joe.0.1740267

I. R. Schlaepfer, L. Rider, L. U. Rodrigues, M. A. Gijon, C. T. Pac et al., Lipid Catabolism via CPT1 as a Therapeutic Target for Prostate Cancer, Mol Cancer Ther, vol.13, issue.10, pp.2361-71, 2014.

J. Chen, B. M. Enloe, P. Weybright, N. Campbell, D. Dorfman et al., Biochemical correlates of thiazolidinedione-induced adipocyte differentiation by highresolution magic angle spinning NMR spectroscopy, Magn Reson Med, vol.48, issue.4, pp.602-612, 2002.

D. 'amato, V. Raimondo, L. Formisano, L. Giuliano, M. et al., Mechanisms of lapatinib resistance in HER2-driven breast cancer, Cancer Treat Rev, vol.41, issue.10, pp.877-83, 2015.

T. Trowe, S. Boukouvala, K. Calkins, R. E. Cutler, R. Fong et al., EXEL-7647 Inhibits Mutant Forms of ErbB2 Associated with Lapatinib Resistance and Neoplastic Transformation, Clin Cancer Res, vol.14, issue.8, pp.2465-75, 2008.

T. R. Wilson, J. Fridlyand, Y. Yan, E. Penuel, L. Burton et al., Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors, Nature, vol.487, issue.7408, pp.505-514, 2012.
DOI : 10.1038/nature11249

URL : http://europepmc.org/articles/pmc3724525?pdf=render

L. A. Gilbert and M. T. Hemann, DNA damage-mediated induction of a chemoresistant niche, Cell, vol.143, issue.3, pp.355-66, 2010.

B. G. Gabrielsson, J. M. Johansson, E. Jennische, M. Jernås, Y. Itoh et al., Depotspecific expression of fibroblast growth factors in human adipose tissue, Obes Res, vol.10, issue.7, pp.608-624, 2002.

L. N. Bell, L. Cai, B. H. Johnstone, D. O. Traktuev, K. L. March et al., A central role for hepatocyte growth factor in adipose tissue angiogenesis, Am J Physiol Endocrinol Metab, vol.294, issue.2, pp.336-344, 2008.

S. Madec, M. Chiarugi, E. Santini, C. Rossi, P. Miccoli et al., Pattern of expression of inflammatory markers in adipose tissue of untreated hypertensive patients, J Hypertens, vol.28, issue.7, pp.1459-65, 2010.

, WHO | Obesity and overweight

K. H. Schmitz, M. L. Neuhouser, T. Agurs-collins, K. A. Zanetti, L. Cadmus-bertram et al., Impact of obesity on cancer survivorship and the potential relevance of race and ethnicity, J Natl Cancer Inst. 18 sept, vol.105, issue.18, p.134454, 2013.

E. E. Calle, C. Rodriguez, K. Walker-thurmond, and M. J. Thun, Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults, N Engl J Med. 24 avr, vol.348, issue.17, p.162538, 2003.

M. Ewertz, M. Jensen, K. Á. Gunnarsdóttir, I. Højris, E. H. Jakobsen et al., Effect of obesity on prognosis after early-stage breast cancer, J Clin Oncol Off J Am Soc Clin Oncol. 1 janv, vol.29, issue.1, p.2531, 2011.

L. Bochet, A. Meulle, S. Imbert, B. Salles, P. Valet et al., Cancer-associated adipocytes promotes breast tumor radioresistance, Biochem Biophys Res Commun. 22 juill, vol.411, issue.1, p.102, 2011.
DOI : 10.1016/j.bbrc.2011.06.101

URL : https://hal.archives-ouvertes.fr/inserm-00617636

M. N. Duong, A. Cleret, E. Matera, K. Chettab, D. Mathé et al.,

, Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibodydependent cellular cytotoxicity, Breast Cancer Res BCR. 24 avr, vol.17, issue.1, p.57, 2015.

D. Angel, R. E. Blando, J. M. Hogan, M. G. Sandoval, M. A. Lansakara et al.,

T. A. Lutz and S. C. Woods, Overview of Animal Models of Obesity, Curr Protoc Pharmacol Editor Board SJ Enna Ed-Chief Al. sept, vol.CHAPTER, 2012.

M. Goodrick, S. Rawesh, A. Katz, D. R. Miles, J. M. Yudkin et al.,

, Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha, in vivo, J Clin Endocrinol Metab. déc, vol.82, issue.12, p.4196200, 1997.

J. M. Bruun, S. B. Pedersen, and B. Richelsen, Regulation of interleukin 8 production and gene expression in human adipose tissue in vitro, J Clin Endocrinol Metab. mars, vol.86, issue.3, p.126773, 2001.

P. E. Scherer, S. Williams, M. Fogliano, G. Baldini, and H. F. Lodish, A novel serum protein similar to C1q, produced exclusively in adipocytes, J Biol Chem, vol.270, issue.45, p.267469, 1995.

N. Ouchi, J. L. Parker, J. J. Lugus, and K. Walsh, Adipokines in inflammation and metabolic disease, Nat Rev Immunol. févr, vol.11, issue.2, p.8597, 2011.

V. Ablamunits, S. Klebanov, S. Y. Giese, and K. C. Herold, Functional human to mouse adipose tissue xenotransplantation, J Endocrinol. janv, vol.212, issue.1, p.417, 2012.

J. W. Behan, J. P. Yun, M. P. Proektor, E. A. Ehsanipour, A. Arutyunyan et al.,

, Adipocytes impair leukemia treatment in mice, Cancer Res, vol.69, p.786774, 2009.

E. A. Ehsanipour, X. Sheng, J. W. Behan, X. Wang, A. Butturini et al., Adipocytes cause leukemia cell resistance to L-asparaginase via release of glutamine, Cancer Res. 15 mai, vol.73, issue.10, 2013.

P. Malvi, B. Chaube, S. V. Singh, N. Mohammad, V. Pandey et al., Weight control interventions improve therapeutic efficacy of dacarbazine in melanoma by reversing obesity-induced drug resistance, Cancer Metab, vol.4, p.21, 2016.

, JCI-Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice

. Disponible,

P. C. Baer, Adipose-derived mesenchymal stromal/stem cells: An update on their phenotype in vivo and in vitro, World J Stem Cells. 26 juill, vol.6, issue.3, 2014.

, Primer sequences used for real-time qPCR. ADRP, Adipose Differentiation-Related Protein; FABP4, FA Binding Protein 4; HIF1 , Hypoxia Inducible Factor 1-? REFERENCES BIBLIOGRAPHIQUES, vol.1

, WHO | Obesity and overweight

. Disponible,

D. Angelantonio, E. Bhupathiraju, S. Wormser, D. Gao, P. Kaptoge et al., Body-mass index and all-cause mortality: individual-participantdata meta-analysis of 239 prospective studies in four continents, Lancet Lond Engl, vol.20, p.77686, 2016.

B. Lauby-secretan, C. Scoccianti, D. Loomis, Y. Grosse, F. Bianchini et al., Body Fatness and Cancer-Viewpoint of the IARC Working Group, N Engl J Med. 25 août, vol.375, issue.8, p.7948, 2016.

H. Vainio, R. Kaaks, and F. Bianchini, Weight control and physical activity in cancer prevention: international evaluation of the evidence, Eur J Cancer Prev Off J Eur Cancer Prev Organ ECP. août, vol.11, issue.2, pp.94-100, 2002.

M. Arnold, M. Touillaud, L. Dossus, H. Freisling, F. Bray et al., Cancers in France in 2015 attributable to high body mass index, Cancer Epidemiol. févr, vol.52, p.159, 2018.

E. E. Calle, C. Rodriguez, K. Walker-thurmond, and M. J. Thun, Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults, N Engl J Med. 24 avr, vol.348, issue.17, p.162538, 2003.

K. H. Schmitz, M. L. Neuhouser, T. Agurs-collins, K. A. Zanetti, L. Cadmus-bertram et al., Impact of obesity on cancer survivorship and the potential relevance of race and ethnicity, J Natl Cancer Inst. 18 sept, vol.105, issue.18, p.134454, 2013.

, Worldwide cancer statistics

C. Research and U. K. , , 2015.

, SEER Cancer Statistics Review 1975-2007-Previous Version-SEER Cancer Statistics

. Disponible, Breast cancer statistics | World Cancer Research Fund International

. Disponible,

A. Sudhakar, History of Cancer, Ancient and Modern Treatment Methods, J Cancer Sci Ther. 1 déc, vol.1, issue.2, p.14, 2009.

A. G. Hall and M. J. Tilby, Mechanisms of action of, and modes of resistance to, alkylating agents used in the treatment of haematological malignancies, Blood Rev. sept, vol.6, issue.3, 1992.

E. Mini, S. Nobili, B. Caciagli, I. Landini, and T. Mazzei, Cellular pharmacology of gemcitabine, Ann Oncol Off J Eur Soc Med Oncol. mai, vol.17, issue.5, pp.7-12, 2006.

W. B. Parker, Enzymology of purine and pyrimidine antimetabolites used in the treatment of cancer, Chem Rev. juill, vol.109, issue.7, p.288093, 2009.

C. Dumontet and M. A. Jordan, Microtubule-binding agents: a dynamic field of cancer therapeutics, Nat Rev Drug Discov, vol.9, issue.10, p.790803, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00526519

A. Howell and M. Dowsett, Endocrinology and hormone therapy in breast cancer: Aromatase inhibitors versus antioestrogens, Breast Cancer Res, vol.6, issue.6, p.26974, 2004.

M. K. Brawer, Hormonal Therapy for Prostate Cancer, Rev Urol, vol.8, issue.2, p.3547, 2006.

R. Munagala, F. Aqil, and R. C. Gupta, Promising molecular targeted therapies in breast cancer, Indian J Pharmacol. mai, vol.43, issue.3, p.23645, 2011.

D. Chen, M. Frezza, S. Schmitt, J. Kanwar, and Q. P. Dou, Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives, Curr Cancer Drug Targets. mars, vol.11, issue.3, p.23953, 2011.
DOI : 10.2174/156800911794519752

URL : http://europepmc.org/articles/pmc3306611?pdf=render

, IARC Publications-Pathology and Genetics of Tumours of the Breast and Female Genital Organs

. Disponible,

F. M. Blows, K. E. Driver, M. K. Schmidt, A. Broeks, F. E. Van-leeuwen et al., Subtyping of breast cancer by immunohistochemistry to investigate a relationship between subtype and short and long term survival: a collaborative analysis of data for 10,159 cases from 12 studies, PLoS Med. 25 mai, vol.7, issue.5, p.1000279, 2010.

C. A. Parise and V. Caggiano, Breast Cancer Survival Defined by the ER/PR/HER2 Subtypes and a Surrogate Classification according to Tumor Grade and Immunohistochemical Biomarkers, J Cancer Epidemiol, p.469251, 2014.

M. M. Moasser, The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis, Oncogene, vol.26, issue.45, p.646987, 2007.

A. B. Tuck, M. Park, E. E. Sterns, A. Boag, and B. E. Elliott, Coexpression of hepatocyte growth factor and receptor (Met) in human breast carcinoma, Am J Pathol. janv, vol.148, issue.1, p.22532, 1996.

G. Valabrega, F. Montemurro, and M. Aglietta, Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer, Ann Oncol Off J Eur Soc Med Oncol. juin, vol.18, issue.6, p.97784, 2007.

N. Harbeck, M. W. Beckmann, A. Rody, A. Schneeweiss, V. Müller et al., HER2 Dimerization Inhibitor Pertuzumab-Mode of Action and Clinical Data in Breast Cancer, Breast Care Basel Switz. mars, vol.8, issue.1, p.4955, 2013.

M. Barok, H. Joensuu, and J. Isola, Trastuzumab emtansine: mechanisms of action and drug resistance, Breast Cancer Res BCR. 5 mars, vol.16, issue.2, p.209, 2014.
DOI : 10.1186/bcr3621

URL : https://breast-cancer-research.biomedcentral.com/track/pdf/10.1186/bcr3621

W. Jeong, J. H. Doroshow, and S. Kummar, US FDA Approved Oral Kinase Inhibitors for the Treatment of Malignancies, Curr Probl Cancer, vol.37, issue.3, p.11044, 2013.

R. H. Alvarez, V. Valero, and G. N. Hortobagyi, Emerging targeted therapies for breast cancer, J Clin Oncol Off J Am Soc Clin Oncol. 10 juill, vol.28, issue.20, p.336679, 2010.
DOI : 10.1200/jco.2009.25.4011

F. Solca, G. Dahl, A. Zoephel, G. Bader, M. Sanderson et al., Target binding properties and cellular activity of afatinib (BIBW 2992), an irreversible ErbB family blocker, J Pharmacol Exp Ther, vol.343, issue.2, p.34250, 2012.

K. Gunzer, F. Joly, J. Ferrero, J. Gligorov, H. De-mont-serrat et al., A phase II study of afatinib, an irreversible ErbB family blocker, added to letrozole in patients with estrogen receptor-positive hormone-refractory metastatic breast cancer progressing on letrozole, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01259837

W. Xia, R. J. Mullin, B. R. Keith, L. Liu, H. Ma et al., Anti-tumor activity of GW572016: a dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways, Oncogene. 12 sept, vol.21, issue.41, p.625563, 2002.

O. 'neill, F. Madden, S. F. Aherne, S. T. Clynes, M. Crown et al., Gene expression changes as markers of early lapatinib response in a panel of breast cancer cell lines. Mol Cancer, 18 juin, vol.11, p.41, 2012.

L. Tang, Y. Wang, A. Strom, J. Gustafsson, and X. Guan, Lapatinib induces p27(Kip1)-dependent G arrest through both transcriptional and post-translational mechanisms, Cell Cycle Georget Tex. 15 août, vol.12, issue.16, p.266574, 2013.
DOI : 10.4161/cc.25728

URL : http://europepmc.org/articles/pmc3865056?pdf=render

C. Holohan, S. Van-schaeybroeck, D. B. Longley, and P. G. Johnston, Cancer drug resistance: an evolving paradigm, Nat Rev Cancer, vol.13, issue.10, p.71426, 2013.
DOI : 10.1038/nrc3599

D. W. Bell, I. Gore, R. A. Okimoto, N. Godin-heymann, R. Sordella et al., Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR, Nat Genet. déc, vol.37, issue.12, p.13156, 2005.

S. Kobayashi, T. J. Boggon, T. Dayaram, P. A. Jänne, O. Kocher et al., EGFR mutation and resistance of non-small-cell lung cancer to gefitinib, N Engl J Med. 24 févr, vol.352, issue.8, p.78692, 2005.

W. Pao, V. A. Miller, K. A. Politi, G. J. Riely, R. Somwar et al., Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain, PLoS Med. mars, vol.2, issue.3, p.73, 2005.

L. A. Doyle and D. D. Ross, Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene, vol.22, p.734058, 2003.

M. N. Duong, A. Cleret, E. Matera, K. Chettab, D. Mathé et al., Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity, Breast Cancer Res BCR. 24 avr, vol.17, issue.1, p.57, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01820280

D. L. Wheeler, S. Huang, T. J. Kruser, M. M. Nechrebecki, E. A. Armstrong et al., Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members, Oncogene. 26 juin, vol.27, issue.28, p.394456, 2008.

N. V. Sergina, M. Rausch, D. Wang, J. Blair, B. Hann et al., Escape from HERfamily tyrosine kinase inhibitor therapy by the kinase-inactive HER3, Nature. 25 janv, vol.445, issue.7126, p.43741, 2007.

D. 'amato, V. Raimondo, L. Formisano, L. Giuliano, M. et al., Mechanisms of lapatinib resistance in HER2-driven breast cancer, Cancer Treat Rev. déc, vol.41, issue.10, p.87783, 2015.

L. V. Sequist, B. A. Waltman, D. Dias-santagata, S. Digumarthy, A. B. Turke et al., Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors, Sci Transl Med. 23 mars, vol.3, issue.75, pp.75-101, 2011.
DOI : 10.1126/scitranslmed.3002003

URL : http://stm.sciencemag.org/content/scitransmed/3/75/75ra26.full.pdf

H. Uramoto, H. Shimokawa, T. Hanagiri, M. Kuwano, and M. Ono, Expression of selected gene for acquired drug resistance to EGFR-TKI in lung adenocarcinoma, Lung Cancer Amst Neth. sept, vol.73, issue.3, p.3615, 2011.

D. W. Mcmillin, J. M. Negri, and C. S. Mitsiades, The role of tumour-stromal interactions in modifying drug response: challenges and opportunities, Nat Rev Drug Discov. mars, vol.12, issue.3, p.21728, 2013.

E. Danen, Integrins: regulators of tissue function and cancer progression, Curr Pharm Des, vol.11, issue.7, p.88191, 2005.

D. Lesniak, Y. Xu, J. Deschenes, R. Lai, J. Thoms et al., Beta1-integrin circumvents the antiproliferative effects of trastuzumab in human epidermal growth factor receptor-2positive breast cancer, Cancer Res, vol.69, issue.22, p.86208, 2009.

T. R. Wilson, J. Fridlyand, Y. Yan, E. Penuel, L. Burton et al., Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors, Nature. 26 juill, vol.487, issue.7408, p.5059, 2012.
DOI : 10.1038/nature11249

URL : http://europepmc.org/articles/pmc3724525?pdf=render

L. A. Gilbert and M. T. Hemann, DNA damage-mediated induction of a chemoresistant niche, Cell, vol.29, issue.3, p.35566, 2010.

S. Cinti, Adipose tissues and obesity, Ital J Anat Embryol Arch Ital Anat Ed Embriologia. juin, vol.104, issue.2, p.3751, 1999.

A. Giordano, A. Smorlesi, A. Frontini, G. Barbatelli, and C. S. White, brown and pink adipocytes: the extraordinary plasticity of the adipose organ, Eur J Endocrinol. mai, vol.170, issue.5, pp.159-171, 2014.

A. Giordano, A. Frontini, and S. Cinti, Convertible visceral fat as a therapeutic target to curb obesity, Nat Rev Drug Discov, vol.15, issue.6, p.40524, 2016.
DOI : 10.1038/nrd.2016.31

S. Cinti, The adipose organ at a glance, Dis Model Mech. sept, vol.5, issue.5, p.58894, 2012.

J. M. Ntambi and K. Young-cheul, Adipocyte Differentiation and Gene Expression, J Nutr. 12 janv, vol.130, issue.12, pp.3122-3126, 2000.

J. M. Moreno-navarrete and J. M. Fernández-real, Adipocyte Differentiation, Adipose Tissue Biology

, , p.1738, 2012.

N. Fukuda and J. A. Ontko, Interactions between fatty acid synthesis, oxidation, and esterification in the production of triglyceride-rich lipoproteins by the liver, J Lipid Res. août, vol.25, issue.8, p.83142, 1984.

P. A. Mayes and D. L. Topping, Regulation of hepatic lipogenesis by plasma free fatty acids: simultaneous studies on lipoprotein secretion, cholesterol synthesis, ketogenesis and gluconeogenesis, Biochem J. avr, vol.140, issue.1, p.1114, 1974.

V. Large, S. Reynisdottir, D. Langin, K. Fredby, M. Klannemark et al., Decreased expression and function of adipocyte hormone-sensitive lipase in subcutaneous fat cells of obese subjects, J Lipid Res, vol.40, issue.11, p.205966, 1999.
URL : https://hal.archives-ouvertes.fr/hal-01983641

S. Reynisdottir, D. Langin, K. Carlström, C. Holm, S. Rössner et al., Effects of weight reduction on the regulation of lipolysis in adipocytes of women with upper-body obesity, Clin Sci Lond Engl, vol.89, issue.4, p.4219, 1979.

G. Haemmerle, R. Zimmermann, M. Hayn, C. Theussl, G. Waeg et al., Hormonesensitive lipase deficiency in mice causes diglyceride accumulation in adipose tissue, muscle, and testis, J Biol Chem. 15 févr, vol.277, issue.7, p.480615, 2002.

J. Osuga, S. Ishibashi, T. Oka, H. Yagyu, R. Tozawa et al., Targeted disruption of hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity, Proc Natl Acad Sci, vol.18, issue.2, p.78792, 2000.

J. A. Villena, S. Roy, E. Sarkadi-nagy, K. Kim, and H. S. Sul, Desnutrin, an adipocyte gene encoding a novel patatin domain-containing protein, is induced by fasting and glucocorticoids: ectopic expression of desnutrin increases triglyceride hydrolysis, J Biol Chem, vol.279, issue.45, p.4706675, 2004.

R. Zimmermann, G. Haemmerle, E. M. Wagner, J. G. Strauss, D. Kratky et al., Decreased fatty acid esterification compensates for the reduced lipolytic activity in hormone-sensitive lipase-deficient white adipose tissue, J Lipid Res, vol.44, issue.11, p.208999, 2003.

K. G. Soni, R. Lehner, P. Metalnikov, O. Donnell, P. Semache et al., Carboxylesterase 3 (EC 3.1.1.1) is a major adipocyte lipase, J Biol Chem. 24 sept, vol.279, issue.39, p.406839, 2004.

V. W. Dolinsky, D. Gilham, G. M. Hatch, L. B. Agellon, R. Lehner et al., Regulation of triacylglycerol hydrolase expression by dietary fatty acids and peroxisomal proliferatoractivated receptors, Biochim Biophys Acta, vol.1635, issue.1, p.208, 2003.

E. Wei, W. Gao, and R. Lehner, Attenuation of adipocyte triacylglycerol hydrolase activity decreases basal fatty acid efflux, J Biol Chem. 16 mars, vol.282, issue.11, p.802735, 2007.

S. Baulande, F. Lasnier, M. Lucas, and J. Pairault, Adiponutrin, a transmembrane protein corresponding to a novel dietary-and obesity-linked mRNA specifically expressed in the adipose lineage, J Biol Chem. 7 sept, vol.276, issue.36, p.3333644, 2001.

C. M. Jenkins, D. J. Mancuso, W. Yan, H. F. Sims, B. Gibson et al., Identification, cloning, expression, and purification of three novel human calcium-independent phospholipase A2 family members possessing triacylglycerol lipase and acylglycerol transacylase activities, J Biol Chem, vol.279, issue.47, p.4896875, 2004.

H. Tornqvist and P. Belfrage, Purification and some properties of a monoacylglycerolhydrolyzing enzyme of rat adipose tissue, J Biol Chem. 10 févr, vol.251, issue.3, p.8139, 1976.

A. S. Greenberg, J. J. Egan, S. A. Wek, N. B. Garty, E. J. Blanchette-mackie et al., Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets, J Biol Chem. 15 juin, vol.266, issue.17, p.113416, 1991.

P. Liu, Y. Ying, Y. Zhao, D. I. Mundy, M. Zhu et al., Chinese hamster ovary K2 cell lipid droplets appear to be metabolic organelles involved in membrane traffic, J Biol Chem. 30 janv, vol.279, issue.5, p.378792, 2004.

A. S. Greenberg, J. J. Egan, S. A. Wek, M. C. Moos, C. Londos et al., Isolation of cDNAs for perilipins A and B: sequence and expression of lipid droplet-associated proteins of adipocytes, Proc Natl Acad Sci, vol.90, issue.24, p.120359, 1993.

J. T. Tansey, C. Sztalryd, J. Gruia-gray, D. L. Roush, J. V. Zee et al., Perilipin ablation results in a lean mouse with aberrant adipocyte lipolysis, enhanced leptin production, and resistance to diet-induced obesity, Proc Natl Acad Sci U S A. 22 mai, vol.98, issue.11, p.64949, 2001.

D. L. Brasaemle, B. Rubin, I. A. Harten, J. Gruia-gray, A. R. Kimmel et al., Perilipin A increases triacylglycerol storage by decreasing the rate of triacylglycerol hydrolysis, J Biol Chem. 8 déc, vol.275, issue.49, p.3848693, 2000.

H. Miyoshi, S. C. Souza, H. Zhang, K. J. Strissel, M. A. Christoffolete et al., Perilipin promotes hormone-sensitive lipase-mediated adipocyte lipolysis via phosphorylationdependent and-independent mechanisms, J Biol Chem. 9 juin, vol.281, issue.23, p.1583744, 2006.

N. R. Coe, M. A. Simpson, and D. A. Bernlohr, Targeted disruption of the adipocyte lipid-binding protein (aP2 protein) gene impairs fat cell lipolysis and increases cellular fatty acid levels, J Lipid Res. mai, vol.40, issue.5, p.96772, 1999.

A. W. Cohen, B. Razani, W. Schubert, T. M. Williams, X. B. Wang et al., Role of caveolin-1 in the modulation of lipolysis and lipid droplet formation, Diabetes. mai, vol.53, issue.5, p.126170, 2004.

D. L. Brasaemle, G. Dolios, L. Shapiro, and R. Wang, Proteomic analysis of proteins associated with lipid droplets of basal and lipolytically stimulated 3T3-L1 adipocytes, J Biol Chem, vol.279, issue.45, p.4683542, 2004.

. Duan-y-min, . Yuan-z-fang, J. Yao, T. Zhang, . Ting et al.,

, Beijing Da Xue Xue Bao, vol.43, p.11722, 2011.

L. J. Syu and A. R. Saltiel, Lipotransin: a novel docking protein for hormone-sensitive lipase, Mol Cell. juill, vol.4, issue.1, p.10915, 1999.

J. G. Gao and M. Simon, Molecular screening for GS2 lipase regulators: inhibition of keratinocyte retinylester hydrolysis by TIP47, J Invest Dermatol. sept, vol.126, issue.9, p.208795, 2006.

P. Arner, L. Hellström, H. Wahrenberg, and M. Brönnegård, Beta-adrenoceptor expression in human fat cells from different regions, J Clin Invest, vol.86, issue.5, p.1595600, 1990.

A. Perea, F. Clemente, J. Martinell, M. L. Villanueva-peñacarrillo, and I. Valverde, Physiological effect of glucagon in human isolated adipocytes, Horm Metab Res Horm Stoffwechselforschung Horm Metab. août, vol.27, issue.8, p.3725, 1995.

M. Lafontan, M. Berlan, and C. Carpene, Fat cell adrenoceptors: inter-and intraspecific differences and hormone regulation, Int J Obes, vol.9, issue.1, p.11727, 1985.

T. C. Walther and R. V. Farese, The life of lipid droplets, Biochim Biophys Acta. juin, vol.1791, issue.6, p.45966, 2009.

E. E. Kershaw and J. S. Flier, Adipose tissue as an endocrine organ, J Clin Endocrinol Metab. juin, vol.89, issue.6, p.254856, 2004.

G. Alvarez-llamas, E. Szalowska, M. P. Vries, . De, D. Weening et al., Characterization of the Human Visceral Adipose Tissue Secretome, Mol Cell Proteomics. 4 janv, vol.6, issue.4, p.589600, 2007.

J. M. Friedman and J. L. Halaas, Leptin and the regulation of body weight in mammals, Nature, vol.22, issue.6704, p.76370, 1998.

S. Margetic, C. Gazzola, G. G. Pegg, and R. A. Hill, Leptin: a review of its peripheral actions and interactions, Int J Obes Relat Metab Disord J Int Assoc Study Obes, vol.26, issue.11, pp.1407-1440, 2002.

P. E. Scherer, S. Williams, M. Fogliano, G. Baldini, and H. F. Lodish, A novel serum protein similar to C1q, produced exclusively in adipocytes, J Biol Chem, vol.270, issue.45, p.267469, 1995.

Y. Zhang, R. Proenca, M. Maffei, M. Barone, L. Leopold et al., Positional cloning of the mouse obese gene and its human homologue, Nature. 1 déc, vol.372, issue.6505, p.42532, 1994.

S. C. Chua, W. K. Chung, X. S. Wu-peng, Y. Zhang, S. M. Liu et al., Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor, Science. 16 févr, vol.271, p.9946, 1996.

S. H. Bates, R. N. Kulkarni, M. Seifert, and M. G. Myers, Roles for leptin receptor/STAT3-dependent and-independent signals in the regulation of glucose homeostasis, Cell Metab. mars, vol.1, issue.3, p.16978, 2005.

S. H. Bates, W. H. Stearns, T. A. Dundon, M. Schubert, A. Tso et al., STAT3 signalling is required for leptin regulation of energy balance but not reproduction, Nature. 20 févr, vol.421, issue.6925, p.8569, 2003.

G. S. Hotamisligil, P. Arner, J. F. Caro, R. L. Atkinson, and B. M. Spiegelman, Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance, J Clin Invest. mai, vol.95, issue.5, p.240915, 1995.

J. Ventre, T. Doebber, M. Wu, K. Macnaul, K. Stevens et al., Targeted disruption of the tumor necrosis factor-alpha gene: metabolic consequences in obese and nonobese mice, Diabetes. sept, vol.46, issue.9, p.152631, 1997.

B. Vozarova, C. Weyer, K. Hanson, P. A. Tataranni, C. Bogardus et al., Circulating interleukin-6 in relation to adiposity, insulin action, and insulin secretion, Obes Res. juill, vol.9, issue.7, p.4147, 2001.
DOI : 10.1038/oby.2001.54

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1038/oby.2001.54

A. S. Greenberg, R. P. Nordan, J. Mcintosh, J. C. Calvo, R. O. Scow et al., Interleukin 6 reduces lipoprotein lipase activity in adipose tissue of mice in vivo and in 3T3-L1 adipocytes: a possible role for interleukin 6 in cancer cachexia, Cancer Res. 1 août, vol.52, issue.15, p.41136, 1992.

C. Bélanger, V. Luu-the, P. Dupont, and A. Tchernof, Adipose tissue intracrinology: potential importance of local androgen/estrogen metabolism in the regulation of adiposity, Horm Metab Res Horm Stoffwechselforschung Horm Metab. déc, vol.34, p.73745, 1112.

A. Meseguer, C. Puche, and A. Cabero, Sex steroid biosynthesis in white adipose tissue, Horm Metab Res Horm Stoffwechselforschung Horm Metab. déc, vol.34, p.7316, 1112.
DOI : 10.1055/s-2002-38249

H. Lee, J. K. Lee, and B. Cho, The role of androgen in the adipose tissue of males, World J Mens Health. août, vol.31, issue.2, p.13640, 2013.

. Oms-|-surpoids and . Obésité,

. Disponible,

R. K. Singh, P. Kumar, and K. Mahalingam, Molecular genetics of human obesity: A comprehensive review, C R Biol. févr, vol.340, issue.2, p.87108, 2017.

M. Rydén, On the origin of human adipocytes and the contribution of bone marrow-derived cells, Adipocyte. sept, vol.5, issue.3, p.3127, 2016.

C. Farnier, S. Krief, M. Blache, F. Diot-dupuy, G. Mory et al., Adipocyte functions are modulated by cell size change: potential involvement of an integrin/ERK signalling pathway, Int J Obes Relat Metab Disord J Int Assoc Study Obes, vol.27, issue.10, p.117886, 2003.
DOI : 10.1038/sj.ijo.0802399

URL : https://www.nature.com/articles/0802399.pdf

J. Boucher, I. Castan-laurell, D. Daviaud, C. Guigné, M. Buléon et al., Adipokine expression profile in adipocytes of different mouse models of obesity, Horm Metab Res Horm Stoffwechselforschung Horm Metab. déc, vol.37, issue.12, p.7617, 2005.

S. P. Weisberg, D. Mccann, M. Desai, M. Rosenbaum, R. L. Leibel et al., Obesity is associated with macrophage accumulation in adipose tissue, J Clin Invest. déc, vol.112, issue.12, p.1796808, 2003.
DOI : 10.1172/jci19246

URL : http://www.jci.org/articles/view/19246/files/pdf

C. R. King, M. T. Spiotto, and D. S. Kapp, Obesity and risk of biochemical failure for patients receiving salvage radiotherapy after prostatectomy, Int J Radiat Oncol Biol Phys. 15 mars, vol.73, issue.4, p.101722, 2009.
DOI : 10.1016/j.ijrobp.2008.06.1118

S. S. Strom, A. M. Kamat, S. K. Gruschkus, Y. Gu, S. Wen et al., Influence of obesity on biochemical and clinical failure after external-beam radiotherapy for localized prostate cancer, Cancer. 1 août, vol.107, issue.3, p.6319, 2006.

H. Geinitz, R. Thamm, T. Mueller, J. K. Zimmermann, F. B. Molls et al., Impact of body mass index on outcomes after conformal radiotherapy in patients with prostate cancer, Int J Radiat Oncol Biol Phys. 1 sept, vol.81, issue.1, p.1622, 2011.

T. Zilli, T. V. Nguyen, J. Bahary, M. Chagnon, A. Dufresne et al., Prognostic impact of abdominal adiposity, waist circumference and body mass index in patients with intermediate-risk prostate cancer treated with radiotherapy, Int J Obes, vol.35, issue.11, p.14216, 2005.

R. Shridhar, T. Hayman, S. E. Hoffe, J. Weber, K. Almhanna et al., Body mass index and survival in esophageal adenocarcinoma treated with chemoradiotherapy followed by esophagectomy, J Gastrointest Surg Off J Soc Surg Aliment Tract. juill, vol.16, issue.7, pp.1296-302, 2012.

C. C. Lerro, K. A. Mcglynn, and M. B. Cook, A systematic review and meta-analysis of the relationship between body size and testicular cancer, Br J Cancer, vol.26, issue.9, pp.1467-74, 2010.

S. Jiralerspong, E. S. Kim, W. Dong, L. Feng, G. N. Hortobagyi et al., Obesity, diabetes, and survival outcomes in a large cohort of early-stage breast cancer patients, Ann Oncol Off J Eur Soc Med Oncol. oct, vol.24, issue.10, p.250614, 2013.

N. Parekh, U. Chandran, and E. V. Bandera, Obesity in cancer survival, Annu Rev Nutr. 21 août, vol.32, p.31142, 2012.

M. Ewertz, M. Jensen, K. Á. Gunnarsdóttir, I. Højris, E. H. Jakobsen et al., Effect of obesity on prognosis after early-stage breast cancer, J Clin Oncol Off J Am Soc Clin Oncol. 1 janv, vol.29, issue.1, p.2531, 2011.

J. A. Crozier, A. Moreno-aspitia, K. V. Ballman, A. C. Dueck, B. A. Pockaj et al., Effect of body mass index on tumor characteristics and disease-free survival in patients from the HER2-positive adjuvant trastuzumab trial N9831, Cancer. 1 juill, vol.119, issue.13, p.244754, 2013.

L. Simkens, M. Koopman, L. Mol, G. J. Veldhuis, T. Bokkel-huinink et al., Influence of body mass index on outcome in advanced colorectal cancer patients receiving chemotherapy with or without targeted therapy, Eur J Cancer Oxf Engl, vol.47, issue.17, p.25607, 1990.

B. Guiu, J. M. Petit, F. Bonnetain, S. Ladoire, S. Guiu et al., Visceral fat area is an independent predictive biomarker of outcome after first-line bevacizumab-based treatment in metastatic colorectal cancer, Gut. mars, vol.59, issue.3, p.3417, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00476649

G. S. Patel, S. Ullah, C. Beeke, P. Hakendorf, R. Padbury et al., Association of BMI with overall survival in patients with mCRC who received chemotherapy versus EGFR and VEGF-targeted therapies, Cancer Med. oct, vol.4, issue.10, p.146171, 2015.

J. J. Griggs and M. S. Sabel, Obesity and cancer treatment: weighing the evidence, J Clin Oncol Off J Am Soc Clin Oncol. 1 sept, vol.26, issue.25, p.40602, 2008.

J. W. Behan, J. P. Yun, M. P. Proektor, E. A. Ehsanipour, A. Arutyunyan et al., Adipocytes impair leukemia treatment in mice, Cancer Res, vol.69, p.786774, 2009.

E. A. Ehsanipour, X. Sheng, J. W. Behan, X. Wang, A. Butturini et al., Adipocytes cause leukemia cell resistance to L-asparaginase via release of glutamine, Cancer Res. 15 mai, vol.73, issue.10, p.29983006, 2013.

P. Malvi, B. Chaube, S. V. Singh, N. Mohammad, V. Pandey et al., Weight control interventions improve therapeutic efficacy of dacarbazine in melanoma by reversing obesity-induced drug resistance, Cancer Metab, vol.4, p.21, 2016.

D. Angel, R. E. Blando, J. M. Hogan, M. G. Sandoval, M. A. Lansakara et al., Stearoyl gemcitabine nanoparticles overcome obesity-induced cancer cell resistance to gemcitabine in a mouse postmenopausal breast cancer model, Cancer Biol Ther. avr, vol.14, issue.4, p.35764, 2013.

Z. Liu, J. Xu, J. He, H. Liu, P. Lin et al., Mature adipocytes in bone marrow protect myeloma cells against chemotherapy through autophagy activation, Oncotarget, vol.6, issue.33, p.34329, 2015.

K. M. Nieman, H. A. Kenny, C. V. Penicka, A. Ladanyi, R. Buell-gutbrod et al., Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth, Nat Med, vol.17, issue.11, p.1498503, 2011.

X. Sheng, J. Tucci, J. Parmentier, J. L. Behan, J. W. Heisterkamp et al., Adipocytes cause leukemia cell resistance to daunorubicin via oxidative stress response, Oncotarget, vol.7, issue.45, p.7314759, 2016.

W. Yu, D. Cao, Q. Li, H. Mei, Y. Hu et al., Adipocytes secreted leptin is a pro-tumor factor for survival of multiple myeloma under chemotherapy, Oncotarget. 27 déc, vol.7, issue.52, p.8607586, 2016.

L. Bochet, A. Meulle, S. Imbert, B. Salles, P. Valet et al., Cancer-associated adipocytes promotes breast tumor radioresistance, Biochem Biophys Res Commun. 22 juill, vol.411, issue.1, p.1026, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00617636

A. Yeung, C. L. Co, N. Tsuruga, T. Yeung, T. Kwan et al., Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1, Nat Commun. 29 mars, vol.7, p.11150, 2016.

J. W. Polli, J. E. Humphreys, K. A. Harmon, S. Castellino, O. Mara et al., (methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine (GW572016, lapatinib) disposition and drug interactions, Drug Metab Dispos Biol Fate Chem. avr, vol.36, issue.4, pp.695-701, 2008.

J. W. Behan, V. I. Avramis, J. P. Yun, S. G. Louie, and S. D. Mittelman, Diet-induced obesity alters vincristine pharmacokinetics in blood and tissues of mice, Pharmacol Res. mai, vol.61, issue.5, p.38590, 2010.

X. Sheng, J. Tucci, J. Behan, and S. D. Mittelman, Abstract 172: Adipocytes decrease daunorubicin concentration in acute lymphoblastic leukemia cells, Cancer Res, vol.74, p.172172, 2014.

G. I. Shapiro and J. W. Harper, Anticancer drug targets: cell cycle and checkpoint control, J Clin Invest. 15 déc, vol.104, issue.12, p.164553, 1999.

J. F. Bromberg, M. H. Wrzeszczynska, G. Devgan, Y. Zhao, R. G. Pestell et al., Stat3 as an oncogene, Cell. 6 août, vol.98, issue.3, p.295303, 1999.

M. Bartucci, S. Svensson, L. Ricci-vitiani, R. Dattilo, M. Biffoni et al., Obesity hormone leptin induces growth and interferes with the cytotoxic effects of 5-fluorouracil in colorectal tumor stem cells, Endocr Relat Cancer. sept, vol.17, issue.3, p.82333, 2010.

Y. Wang, G. Morrison, R. Gillihan, J. Guo, R. M. Ward et al., Different mechanisms for resistance to trastuzumab versus lapatinib in HER2-positive breast cancers-role of estrogen receptor and HER2 reactivation, Breast Cancer Res BCR, vol.13, issue.6, p.121, 2011.

K. N. Frayn, K. Khan, S. W. Coppack, and M. Elia, Amino acid metabolism in human subcutaneous adipose tissue in vivo, Clin Sci Lond Engl, vol.80, issue.5, p.4714, 1979.

M. Simons and G. Raposo, Exosomes-vesicular carriers for intercellular communication, Curr Opin Cell Biol. août, vol.21, issue.4, p.57581, 2009.

M. Scaltriti, F. Rojo, A. Ocaña, J. Anido, M. Guzman et al., Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer, J Natl Cancer Inst, vol.99, issue.8, p.62838, 2007.

K. Tamura, C. Shimizu, T. Hojo, S. Akashi-tanaka, T. Kinoshita et al., Fc?R2A and 3A polymorphisms predict clinical outcome of trastuzumab in both neoadjuvant and metastatic settings in patients with HER2-positive breast cancer, Ann Oncol Off J Eur Soc Med Oncol. juin, vol.22, issue.6, p.13027, 2011.

R. Nahta, T. Takahashi, N. T. Ueno, M. Hung, and F. J. Esteva, P27(kip1) down-regulation is associated with trastuzumab resistance in breast cancer cells, Cancer Res. 1 juin, vol.64, issue.11, p.39816, 2004.

K. Berns, H. M. Horlings, B. T. Hennessy, M. Madiredjo, E. M. Hijmans et al., A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell, vol.12, p.395402, 2007.

L. Formisano, L. Nappi, R. Rosa, R. Marciano, D. 'amato et al., Epidermal growth factor-receptor activation modulates Src-dependent resistance to lapatinib in breast cancer models, Breast Cancer Res BCR. 5 mai, vol.16, issue.3, p.45, 2014.

B. N. Rexer, A. Ham, C. Rinehart, S. Hill, M. Granja-ingram-n-de et al., Phosphoproteomic mass spectrometry profiling links Src family kinases to escape from HER2 tyrosine kinase inhibition, Oncogene, vol.30, issue.40, p.416374, 2011.

D. J. Slamon, G. M. Clark, S. G. Wong, W. J. Levin, A. Ullrich et al., Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene, Science. 9 janv, vol.235, issue.4785, p.17782, 1987.

C. L. Vogel, M. A. Cobleigh, D. Tripathy, J. C. Gutheil, L. N. Harris et al., Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer, J Clin Oncol Off J Am Soc Clin Oncol. 1 févr, vol.20, issue.3, p.71926, 2002.

M. A. Cobleigh, C. L. Vogel, D. Tripathy, N. J. Robert, S. Scholl et al., Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease, J Clin Oncol Off J Am Soc Clin Oncol. sept, vol.17, issue.9, p.263948, 1999.

E. H. Romond, E. A. Perez, J. Bryant, V. J. Suman, C. E. Geyer et al., Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer, N Engl J Med, vol.353, issue.16, p.167384, 2005.

C. E. Geyer, J. Forster, D. Lindquist, S. Chan, C. G. Romieu et al., Lapatinib plus capecitabine for HER2-positive advanced breast cancer, N Engl J Med. 28 déc, vol.355, issue.26, p.273343, 2006.

L. Liu, J. Greger, H. Shi, Y. Liu, J. Greshock et al., Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: activation of AXL, Cancer Res. 1 sept, vol.69, issue.17, p.68718, 2009.

W. Xia, S. Bacus, P. Hegde, I. Husain, J. Strum et al., A model of acquired autoresistance to a potent ErbB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in breast cancer, Proc Natl Acad Sci. 16 mai, vol.103, issue.20, p.7795800, 2006.

J. W. Kim, H. Kim, S. Im, S. Kang, H. S. Hur et al., The growth inhibitory effect of lapatinib, a dual inhibitor of EGFR and HER2 tyrosine kinase, in gastric cancer cell lines, Cancer Lett. 18 déc, vol.272, issue.2, p.296306, 2008.

R. Diaz, P. A. Nguewa, R. Parrondo, C. Perez-stable, I. Manrique et al., Antitumor and antiangiogenic effect of the dual EGFR and HER-2 tyrosine kinase inhibitor lapatinib in a lung cancer model, BMC Cancer. 11 mai, vol.10, p.188, 2010.

P. C. Baer, Adipose-derived mesenchymal stromal/stem cells: An update on their phenotype in vivo and in vitro, World J Stem Cells. 26 juill, vol.6, issue.3, p.25665, 2014.
DOI : 10.4252/wjsc.v6.i3.256

URL : https://doi.org/10.4252/wjsc.v6.i3.256

J. Serr, Y. Suh, and K. Lee, Regulation of adipose triglyceride lipase by fasting and refeeding in avian species, Poult Sci. 1 déc, vol.88, issue.12, p.258591, 2009.

J. Nedvídková, K. Smitka, V. Kopský, and V. Hainer, Adiponectin, an adipocyte-derived protein, Physiol Res, vol.54, issue.2, p.13340, 2005.

J. M. Bruun, S. B. Pedersen, and B. Richelsen, Regulation of interleukin 8 production and gene expression in human adipose tissue in vitro, J Clin Endocrinol Metab. mars, vol.86, issue.3, pp.1267-73, 2001.

B. Dirat, L. Bochet, M. Dabek, D. Daviaud, S. Dauvillier et al., Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion, Cancer Res. 1 avr, vol.71, issue.7, p.245565, 2011.
DOI : 10.1158/0008-5472.can-10-3323

URL : https://hal.archives-ouvertes.fr/inserm-00819288

, Secretome analysis of rat adipose tissues shows location-specific roles for each depot type

. Disponible,

D. Zhang, A. Pal, W. G. Bornmann, F. Yamasaki, F. J. Esteva et al., Activity of lapatinib is independent of EGFR expression level in HER2-overexpressing breast cancer cells, Mol Cancer Ther. juill, vol.7, issue.7, p.184650, 2008.

N. Grabinski and F. Ewald, Ibrutinib (ImbruvicaTM) potently inhibits ErbB receptor phosphorylation and cell viability of ErbB2-positive breast cancer cells, Invest New Drugs. déc, vol.32, issue.6, p.1096104, 2014.

P. R. Gavine, L. Mooney, E. Kilgour, A. P. Thomas, K. Al-kadhimi et al., AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family, Cancer Res. 15 avr, vol.72, issue.8, p.204556, 2012.

I. F. Benter, F. Sarkhou, A. -. Khaldi, A. T. Chandrasekhar, B. Attur et al., The dual targeting of EGFR and ErbB2 with the inhibitor Lapatinib corrects high glucose-induced apoptosis and vascular dysfunction by opposing multiple diabetes-induced signaling changes, J Drug Target, vol.23, issue.6, p.50618, 2015.

D. W. Rusnak, K. Lackey, K. Affleck, E. R. Wood, K. J. Alligood et al., The effects of the novel, reversible epidermal growth factor receptor/ErbB-2 tyrosine kinase inhibitor, GW2016, on the growth of human normal and tumor-derived cell lines in vitro and in vivo, Mol Cancer Ther. déc, vol.1, issue.2, p.8594, 2001.

Z. A. Wainberg, A. Anghel, A. J. Desai, R. Ayala, T. Luo et al., Lapatinib, a dual EGFR and HER2 kinase inhibitor, selectively inhibits HER2-amplified human gastric cancer cells and is synergistic with trastuzumab in vitro and in vivo, Clin Cancer Res Off J Am Assoc Cancer Res. 1 mars, vol.16, issue.5, p.150919, 2010.

G. E. Konecny, M. D. Pegram, N. Venkatesan, R. Finn, G. Yang et al., Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells, Cancer Res. 1 févr, vol.66, issue.3, p.16309, 2006.

S. W. Brady, J. Zhang, M. Tsai, and D. Yu, PI3K-independent mTOR activation promotes lapatinib resistance and IAP expression that can be effectively reversed by mTOR and Hsp90 inhibition, Cancer Biol Ther, vol.16, issue.3, p.40211, 2015.
DOI : 10.1080/15384047.2014.1002693

URL : https://www.tandfonline.com/doi/pdf/10.1080/15384047.2014.1002693?needAccess=true

A. Vazquez-martin, C. Oliveras-ferraros, R. Colomer, J. Brunet, and J. A. Menendez, Low-scale phosphoproteome analyses identify the mTOR effector p70 S6 kinase 1 as a specific biomarker of the dual-HER1/HER2 tyrosine kinase inhibitor lapatinib (Tykerb®) in human breast carcinoma cells, Ann Oncol. 1 juin, vol.19, issue.6, p.1097109, 2008.

V. Ablamunits, S. Klebanov, S. Y. Giese, and K. C. Herold, Functional human to mouse adipose tissue xenotransplantation, J Endocrinol. janv, vol.212, issue.1, p.417, 2012.
DOI : 10.1530/joe-11-0201

URL : https://joe.bioscientifica.com/downloadpdf/journals/joe/212/1/41.pdf

, JCI-Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice

W. Zhu and C. M. Nelson, Adipose and mammary epithelial tissue engineering
DOI : 10.4161/biom.24630

URL : https://www.tandfonline.com/doi/pdf/10.4161/biom.24630?needAccess=true

. Disponible,

V. Laurent, A. Guérard, C. Mazerolles, L. Gonidec, S. Toulet et al., Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity, Nat Commun [Internet], vol.12, issue.2016

L. Merrill, M. Emond, C. Kim, M. J. Antignac, J. et al., Toxicological Function of Adipose Tissue: Focus on Persistent Organic Pollutants, Environ Health Perspect. févr, vol.121, issue.2, p.1629, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01549161

T. L. Whiteside, Targeting adenosine in cancer immunotherapy: a review of recent progress, Expert Rev Anticancer Ther. 3 juin, vol.17, issue.6, p.52735, 2017.

C. Olesch, W. Sha, C. Angioni, L. K. Sha, E. Açaf et al., MPGES-1-derived PGE2 suppresses CD80 expression on tumor-associated phagocytes to inhibit anti-tumor immune responses in breast cancer, Oncotarget, vol.30, issue.2015

M. Majumder, X. Xin, L. Liu, G. V. Girish, and P. K. Lala, Prostaglandin E2 receptor EP4 as the common target on cancer cells and macrophages to abolish angiogenesis, lymphangiogenesis, metastasis, and stem-like cell functions, Cancer Sci. sept, vol.105, issue.9, p.114251, 2014.

I. Ben-batalla, M. Cubas-cordova, F. Udonta, M. Wroblewski, J. S. Waizenegger et al., Cyclooxygenase-2 blockade can improve efficacy of VEGF-targeting drugs, Oncotarget

T. Puig, H. Aguilar, S. Cufí, G. Oliveras, C. Turrado et al., A novel inhibitor of fatty acid synthase shows activity against HER2+ breast cancer xenografts and is active in anti-HER2 drug-resistant cell lines, Breast Cancer Res, vol.13, issue.6

, Anti-obesity effect of sulfated glucosamine by AMPK signal pathway in 3T3-L1 adipocytesScienceDirect

. Disponible,