C. A. Hudis, Trastuzumab-mechanism of action and use in clinical practice, The New England journal of medicine, vol.357, pp.39-51, 2007.

J. Baselga, Annals of oncology : official journal of the European Society for, Medical Oncology / ESMO, vol.21, pp.36-40, 2010.

J. A. Engelman, J. Luo, and L. C. Cantley, The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism, Nature reviews Genetics, vol.7, pp.606-619, 2006.

A. Harle, M. Lion, and N. Lozano, Analysis of PIK3CA exon 9 and 20 mutations in breast cancers using PCR-HRM and PCR-ARMS: correlation with clinicopathological criteria, Oncology reports, vol.29, pp.1043-1052, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00823892

M. C. Mendoza, E. E. Blenis, and J. , The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation, Trends in biochemical sciences, vol.36, pp.320-328, 2011.

A. M. Martelli, G. Tabellini, and D. Bressanin, The emerging multiple roles of nuclear

. Akt, Biochimica et biophysica acta, vol.1823, pp.2168-2178, 2012.

P. Ss and S. W. Kim, Activated Akt signaling pathway in invasive ductal carcinoma of the breast: correlation with HER2 overexpression, Oncology reports, vol.18, pp.139-143, 2007.

H. J. Lin, F. C. Hsieh, H. Song, and J. Lin, Elevated phosphorylation and activation of PDK

, /AKT pathway in human breast cancer, Br J Cancer, vol.93, pp.1372-1381, 2005.

A. F. Baker, T. Dragovich, N. T. Ihle, R. Williams, C. Fenoglio-preiser et al., Stability of phosphoprotein as a biological marker of tumor signaling, Clin Cancer Res, vol.11, pp.4338-4340, 2005.

K. David and H. Juhl, Immunohistochemical Detection of Phosphoproteins and Cancer Pathways

J. W. Mandell, Phosphorylation state-specific antibodies: applications in investigative and diagnostic pathology, The American journal of pathology, vol.163, pp.1687-1698, 2003.

J. W. Mandell, Immunohistochemical assessment of protein phosphorylation state: the dream and the reality, Histochemistry and cell biology, vol.130, pp.465-471, 2008.

S. Menard, S. M. Pupa, M. Campiglio, and E. Tagliabue, Biologic and therapeutic role of HER2 in cancer, Oncogene, vol.22, pp.6570-6578, 2003.

O. Yersal and S. Barutca, Biological subtypes of breast cancer: Prognostic and therapeutic implications, World journal of clinical oncology, vol.5, pp.412-424, 2014.

D. J. Slamon, B. Leyland-jones, and S. Shak, Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2, The New England journal of medicine, vol.344, pp.783-792, 2001.

G. Valabrega, F. Montemurro, and M. Aglietta, Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer, Medical Oncology / ESMO, vol.18, pp.977-984, 2007.

J. J. Gemmete and S. K. Mukherji, Trastuzumab (herceptin), AJNR American journal of neuroradiology, vol.32, pp.1373-1374, 2011.

F. M. Yakes, W. Chinratanalab, C. A. Ritter, W. King, S. Seelig et al., Herceptininduced inhibition of phosphatidylinositol-3 kinase and Akt Is required for antibodymediated effects on p27, cyclin D1, and antitumor action, Cancer Res, vol.62, pp.4132-4141, 2002.

S. K. Mohsin, H. L. Weiss, and M. C. Gutierrez, Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer, Nature clinical practice Oncology, vol.23, pp.269-280, 2005.

L. Arnould, M. Gelly, and F. Penault-llorca, Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism?, Br J Cancer, vol.94, pp.259-267, 2006.

R. A. Clynes, T. L. Towers, L. G. Presta, and J. V. Ravetch, Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets, Nat Med, vol.6, pp.443-446, 2000.

N. L. Spector and K. L. Blackwell, Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer, J Clin Oncol, vol.27, pp.5838-5847, 2009.

M. A. Molina, J. Codony-servat, J. Albanell, F. Rojo, A. J. Baselga et al., Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells, Cancer Res, vol.61, pp.4744-4749, 2001.

, Erk1/2 during routine fixation of primary breast cancer, Breast cancer research : BCR, vol.12, p.76, 2010.

S. Siddiqui and D. L. Rimm, Pre-analytic variables and phospho-specific antibodies: the Achilles heel of immunohistochemistry, Les Biomarqueurs en Oncologie, vol.12, 2010.

J. Ferlay, I. Soerjomataram, M. Ervik, R. Dikshit, S. Eser et al., Cancer Incidence and Mortality Worldwide: IARC CancerBase No.11. Lyon, France: International Agency for Research on Cancer, 2012.

F. Binder-foucard, N. Bossard, P. Delafosse, A. Belot, A. S. Woronoff et al., Cancer incidence and mortality in France over the 1980-2012 period: solid tumors, Rev Epidemiol Sante Publique, vol.62, issue.2, pp.95-108, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01915012

, Collection Etat des lieux et des connaissances, ouvrage collectif édité par l'INCa, 2013.

J. Robert, Signalisation cellulaire et cancer, 2010.

A. Harle, M. Lion, N. Lozano, M. Husson, V. Harter et al., Analysis of PIK3CA exon 9 and 20 mutations in breast cancers using PCR-HRM and PCR-ARMS: correlation with clinicopathological criteria, Oncol Rep, vol.29, issue.3, pp.1043-52, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00823892

M. Cizkova, A. Susini, S. Vacher, G. Cizeron-clairac, C. Andrieu et al., PIK3CA mutation impact on survival in breast cancer patients and in ERalpha, PR and ERBB2-based subgroups, Breast Cancer Res, vol.14, issue.1, p.28, 2012.

P. Liu, H. Cheng, S. Santiago, M. Raeder, F. Zhang et al., Oncogenic PIK3CA-driven mammary tumors frequently recur via PI3K pathway-dependent and PI3K pathwayindependent mechanisms, Nat Med, vol.17, issue.9, pp.1116-1136, 2011.

B. N. Rexer, S. Chanthaphaychith, K. Dahlman, and C. L. Arteaga, Direct inhibition of PI3K in combination with dual HER2 inhibitors is required for optimal antitumor activity in HER2+ breast cancer cells, Breast Cancer Res, vol.16, issue.1, p.9, 2014.

L. H. Saal, K. Holm, M. Maurer, L. Memeo, T. Su et al., PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma, Cancer Res, vol.65, issue.7, pp.2554-2563, 2005.

, Anatomie du sein, cancer du sein, 2010.

, Les traitements des cancers du sein. Collection Guides patients Cancer info, 2013.

, et Lorraine) de prise en charge du cancer du sein, 2012.

A. Gschwind, O. M. Fischer, and A. Ullrich, The discovery of receptor tyrosine kinases: targets for cancer therapy, Nat Rev Cancer, vol.4, issue.5, pp.361-70, 2004.

P. Blume-jensen and T. Hunter, Oncogenic kinase signalling, Nature, vol.411, issue.6835, pp.355-65, 2001.

P. De-cremoux and J. Robert, Cell signalling and cancer: characterisation of therapeutic targets, Pathol Biol, vol.60, issue.4, pp.217-239, 2012.

P. Lahiry, A. Torkamani, N. J. Schork, and R. A. Hegele, Kinase mutations in human disease: interpreting genotype-phenotype relationships, Nat Rev Genet, vol.11, issue.1, pp.60-74, 2010.

M. A. Lemmon and J. Schlessinger, Cell Signaling by Receptor Tyrosine Kinases, Cell, vol.141, issue.7, pp.1117-1151, 2010.

N. E. Hynes and H. A. Lane, ERBB receptors and cancer: the complexity of targeted inhibitors, Nat Rev Cancer, vol.5, issue.5, pp.341-54, 2005.

N. E. Hynes, K. Horsch, M. A. Olayioye, and A. Badache, The ErbB receptor tyrosine family as signal integrators, Endocr Relat Cancer, 2001.

N. E. Hynes and G. Macdonald, ErbB receptors and signaling pathways in cancer, Curr Opin Cell Biol, vol.21, issue.2, pp.177-84, 2009.

N. V. Sergina and M. M. Moasser, The HER family and cancer: emerging molecular mechanisms and therapeutic targets, Trends Mol Med, vol.13, issue.12, pp.527-561, 2007.

P. Hubert,

, Bull Cancer, vol.94, issue.7, pp.137-182, 2007.

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

H. Linardou, I. J. Dahabreh, D. Bafaloukos, P. Kosmidis, and S. Murray, Somatic EGFR mutations and efficacy of tyrosine kinase inhibitors in NSCLC, Nat Rev Clin Oncol, vol.6, issue.6, pp.352-66, 2009.

Y. Samuels and V. E. Velculescu, Oncogenic mutations of PIK3CA in human cancers, Cell Cycle, vol.3, issue.10, pp.1221-1225, 2004.

P. W. Brandt-rauf, M. R. Pincus, and W. P. Carney, The c-erbB-2 protein in oncogenesis: molecular structure to molecular epidemiology, Crit Rev Oncog, vol.5, issue.2-3, pp.313-342, 1994.

N. Iqbal and N. Iqbal, Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications, Mol Biol Int, p.852748, 2014.

L. C. Padhy, C. Shih, D. Cowing, R. Finkelstein, and R. A. Weinberg, Identification of a phosphoprotein specifically induced by the transforming DNA of rat neuroblastomas, Cell, vol.28, issue.4, pp.865-71, 1982.

A. L. Schechter, D. F. Stern, L. Vaidyanathan, S. J. Decker, J. A. Drebin et al., The neu oncogene: an erb-B-related gene encoding a 185,000-Mr tumour antigen, Nature, vol.312, issue.5994, pp.513-519, 1984.

M. Tan and D. Yu, Molecular mechanisms of erbB2-mediated breast cancer chemoresistance

, Adv Exp Med Biol, vol.608, pp.119-148, 2007.

A. M. Abbott, R. Bueno, M. T. Pedrini, J. M. Murray, and R. J. Smith, Insulin-like growth factor I receptor gene structure, J Biol Chem, vol.267, issue.15, pp.10759-63, 1992.

P. F. Christopoulos, P. Msaouel, and M. Koutsilieris, The role of the insulin-like growth factor-1 system in breast cancer, Mol Cancer, vol.14, p.43, 2015.

J. E. Kucab and S. E. Dunn, Role of IGF-1R in mediating breast cancer invasion and metastasis, Breast Dis, vol.17, pp.41-48, 2003.

S. M. Farabaugh, D. N. Boone, and A. V. Lee, Role of IGF1R in Breast Cancer Subtypes, Stemness, and Lineage Differentiation, Front Endocrinol, vol.6, p.59, 2015.

J. A. Engelman, J. Luo, and L. C. Cantley, The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism, Nat Rev Genet, vol.7, issue.8, pp.606-625, 2006.

M. C. Mendoza, E. E. Er, and J. Blenis, The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation, Trends Biochem Sci, vol.36, issue.6, pp.320-328, 2011.

K. S. Saini, S. Loi, E. De-azambuja, O. Metzger-filho, M. L. Saini et al., Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer, Cancer Treat Rev, vol.39, issue.8, pp.935-981, 2013.

E. Castellano and J. Downward, RAS Interaction with PI3K: More Than Just Another Effector Pathway, Genes Cancer, vol.2, issue.3, pp.261-74, 2011.

A. Carracedo, J. Baselga, and P. P. Pandolfi, Deconstructing feedback-signaling networks to improve anticancer therapy with mTORC1 inhibitors, Cell Cycle, vol.7, issue.24, pp.3805-3814, 2008.

A. Carracedo, L. Ma, J. Teruya-feldstein, F. Rojo, L. Salmena et al., Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer, J Clin Invest, vol.118, issue.9, pp.3065-74, 2008.

P. F. Mcauliffe, F. Meric-bernstam, G. B. Mills, and A. M. Gonzalez-angulo, Deciphering the role of PI3K/Akt/mTOR pathway in breast cancer biology and pathogenesis, Clin Breast Cancer, vol.10, issue.3, pp.59-65, 2010.

C. Brunner-kubath, W. Shabbir, V. Saferding, R. Wagner, C. F. Singer et al., The PI3 kinase/mTOR blocker NVP-BEZ235 overrides resistance against irreversible ErbB inhibitors in breast cancer cells, Breast Cancer Res Treat, vol.129, issue.2, pp.387-400, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00615384

A. M. Gonzalez-angulo, G. R. Blumenschein, and J. , Defining biomarkers to predict sensitivity to PI3K/Akt/mTOR pathway inhibitors in breast cancer, Cancer Treat Rev, vol.39, issue.4, pp.313-333, 2013.

P. Liu, H. Cheng, T. M. Roberts, and J. J. Zhao, Targeting the phosphoinositide 3-kinase pathway in cancer, Nat Rev Drug Discov, vol.8, issue.8, pp.627-671, 2009.

J. C. Bendell, J. Rodon, H. A. Burris, M. De-jonge, J. Verweij et al., Phase I, doseescalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors, J Clin Oncol, vol.30, issue.3, pp.282-90, 2012.

L. Coutte, C. Dreyer, M. P. Sablin, S. Faivre, and R. E. ,

, Bull Cancer, vol.99, issue.2, pp.173-80, 2012.

R. L. Dillon, D. E. White, and W. J. Muller, The phosphatidyl inositol 3-kinase signaling network: implications for human breast cancer, Oncogene, vol.26, issue.9, pp.1338-1383, 2007.

C. A. Castaneda, H. Cortes-funes, H. L. Gomez, and E. M. Ciruelos, The phosphatidyl inositol 3-kinase/AKT signaling pathway in breast cancer, Cancer Metastasis Rev, vol.29, issue.4, pp.751-760, 2010.

P. L. Depowski, S. I. Rosenthal, and J. S. Ross, Loss of expression of the PTEN gene protein product is associated with poor outcome in breast cancer, Mod Pathol, vol.14, issue.7, pp.672-678, 2001.

M. C. Hollander, G. M. Blumenthal, and P. A. Dennis, PTEN loss in the continuum of common cancers, rare syndromes and mouse models, Nat Rev Cancer, vol.11, issue.4, pp.289-301, 2011.

M. Barbareschi, F. Buttitta, L. Felicioni, S. Cotrupi, F. Barassi et al., Different prognostic roles of mutations in the helical and kinase domains of the PIK3CA gene in breast carcinomas, Clin Cancer Res, vol.13, issue.20, pp.6064-6073, 2007.

S. Boyault, Y. Drouet, C. Navarro, T. Bachelot, C. Lasset et al., Mutational characterization of individual breast tumors: TP53 and PI3K pathway genes are frequently and distinctively mutated in different subtypes, Breast Cancer Res Treat, vol.132, issue.1, pp.29-39, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01007700

B. Dave, I. Migliaccio, M. C. Gutierrez, M. F. 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, vol.29, issue.2, pp.166-73, 2011.

L. Wang, Q. Zhang, J. Zhang, S. Sun, H. Guo et al., PI3K pathway activation results in low efficacy of both trastuzumab and lapatinib, BMC Cancer, vol.11, p.248, 2011.

Y. Nagata, K. H. Lan, X. Zhou, M. Tan, F. J. Esteva et al., PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients, Cancer Cell, vol.6, issue.2, pp.117-144, 2004.

P. P. Pandolfi, Breast cancer--loss of PTEN predicts resistance to treatment, N Engl J Med, vol.351, issue.22, pp.2337-2345, 2004.

E. Razis, M. Bobos, V. Kotoula, A. G. Eleftheraki, H. P. Kalofonos et al., Evaluation of the association of PIK3CA mutations and PTEN loss with efficacy of trastuzumab therapy in metastatic breast cancer, Breast Cancer Res Treat, vol.128, issue.2, pp.447-56, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00642847

T. Fujita, H. Doihara, K. Kawasaki, D. Takabatake, H. Takahashi et al., PTEN activity could be a predictive marker of trastuzumab efficacy in the treatment of ErbB2-overexpressing breast cancer, Br J Cancer, vol.94, issue.2, pp.247-52, 2006.

P. Kechagioglou, R. M. Papi, X. Provatopoulou, E. Kalogera, E. Papadimitriou et al., Tumor suppressor PTEN in breast cancer: heterozygosity, mutations and protein expression, Anticancer Res, vol.34, issue.3, pp.1387-400, 2014.

J. Yang, P. Cron, V. Thompson, V. M. Good, D. Hess et al., Molecular Mechanism for the Regulation of Protein Kinase B/Akt by Hydrophobic Motif Phosphorylation, Molecular Cell, vol.9, issue.6, pp.1227-1267, 2002.

K. M. Nicholson and N. G. Anderson, The protein kinase B/Akt signalling pathway in human malignancy, Cell Signal, vol.14, issue.5, pp.381-95, 2002.

E. Gonzalez and T. E. Mcgraw, The Akt kinases: isoform specificity in metabolism and cancer. Cell cycle, vol.8, pp.2502-2510, 2009.

B. D. Manning and L. C. Cantley, AKT/PKB signaling: navigating downstream, Cell, vol.129, issue.7, pp.1261-74, 2007.

A. M. Martelli, G. Tabellini, D. Bressanin, A. Ognibene, K. Goto et al., The emerging multiple roles of nuclear Akt, Biochim Biophys Acta, vol.1823, issue.12, pp.2168-78, 2012.

S. S. Park and S. W. Kim, Activated Akt signaling pathway in invasive ductal carcinoma of the breast: correlation with HER2 overexpression, Oncol Rep, vol.18, issue.1, pp.139-182, 2007.

H. J. Lin, F. C. Hsieh, H. Song, and J. Lin, Elevated phosphorylation and activation of PDK-1/AKT pathway in human breast cancer, Br J Cancer, vol.93, issue.12, pp.1372-81, 2005.

M. Laplante and D. M. Sabatini, mTOR signaling at a glance, J Cell Sci, vol.122, pp.3589-94, 2009.

A. Masahiro, E. Blazek, and P. K. Vogt, A role of the kinase mTOR in cellular transformation induced by the oncoproteins P3k and Akt, Proc Natl Acad Sci USA, vol.98, pp.136-177, 2001.

C. Villarreal-garza, J. Cortes, F. Andre, and S. Verma, mTOR inhibitors in the management of hormone receptor-positive breast cancer: the latest evidence and future directions, Ann Oncol, vol.23, issue.10, pp.2526-2561, 2012.

X. Wang and C. G. Proud, mTORC1 signaling: what we still don't know, J Mol Cell Biol, vol.3, issue.4, pp.206-226, 2011.

D. A. Guertin and D. M. Sabatini, Defining the role of mTOR in cancer, Cancer Cell, vol.12, issue.1, pp.9-22, 2007.

C. Nardella, Z. Chen, L. Salmena, A. Carracedo, A. Alimonti et al., Aberrant Rhebmediated mTORC1 activation and Pten haploinsufficiency are cooperative oncogenic events, Genes Dev, vol.22, issue.16, pp.2172-2179, 2008.

J. Avruch, X. Long, Y. Lin, S. Ortiz-vega, J. Rapley et al., Activation of mTORC1 in two steps: Rheb-GTP activation of catalytic function and increased binding of substrates to raptor, Biochem Soc Trans, vol.37, pp.223-229, 2009.

T. B. Huber, G. Walz, and E. W. Kuehn, mTOR and rapamycin in the kidney: signaling and therapeutic implications beyond immunosuppression, Kidney Int, vol.79, issue.5, pp.502-513, 2011.

S. C. Kozma and G. Thomas, p70s6k/p85s6k: mechanism of activation and role in mitogenesis, Semin Cancer Biol, vol.5, issue.4, pp.255-60, 1994.

P. Brennan, J. W. Babbage, G. Thomas, and D. Cantrell, p70(s6k) integrates phosphatidylinositol 3-kinase and rapamycin-regulated signals for E2F regulation in T lymphocytes, Mol Cell Biol, vol.19, pp.4729-4767, 1999.

G. Thomas and M. N. Hall, TOR signalling and control of cell growth, Curr Opin Cell Biol

M. Laser, V. S. Kasi, M. Hamawaki, C. Gt, C. M. Kerr et al., Differential activation of p70 and p85 S6 kinase isoforms during cardiac hypertrophy in the adult mammal, J Biol Chem, vol.273, issue.38, pp.24610-24619, 1998.

M. Mahalingam and D. J. Templeton, Constitutive activation of S6 kinase by deletion of aminoterminal autoinhibitory and rapamycin sensitivity domains, Mol Cell Biol, vol.16, issue.1, pp.405-418, 1996.

Q. P. Weng, M. Kozlowski, C. Belham, A. Zhang, M. J. Comb et al., Regulation of the p70 S6 kinase by phosphorylation in vivo. Analysis using site-specific anti-phosphopeptide antibodies, J Biol Chem, vol.273, issue.26, pp.16621-16630, 1998.

L. A. Berven and M. F. Crouch, Cellular function of p70S6K: A role in regulating cell motility, Immunol Cell Biol, vol.78, issue.4, pp.447-51, 2000.

D. R. Alessi, M. T. Kozlowski, Q. P. Weng, N. Morrice, and J. Avruch, 3-Phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylates and activates the p70 S6 kinase in vivo and in vitro, Curr Biol, vol.8, issue.2, pp.69-81, 1998.

B. W. Doble and J. R. Woodgett, GSK-3: tricks of the trade for a multi-tasking kinase, J Cell Sci, vol.116, pp.1175-86, 2003.

P. Cohen and S. Frame, The renaissance of GSK3, Nat Rev Mol Cell Biol, vol.2, issue.10, pp.769-76, 2001.

A. Cole, S. Frame, and P. Cohen, Further evidence that the tyrosine phosphorylation of glycogen synthase kinase-3 (GSK3) in mammalian cells is an autophosphorylation event, Biochem J, vol.377, pp.249-55, 2004.

U. Beffert, G. Morfini, H. H. Bock, H. Reyna, S. T. Brady et al., Reelin-mediated signaling locally regulates protein kinase B/Akt and glycogen synthase kinase 3beta, J Biol Chem, vol.277, issue.51, pp.49958-64, 2002.

J. A. Diehl, M. Cheng, M. F. Roussel, and C. J. Sherr, Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization, Genes Dev, vol.12, pp.3499-511, 1998.

A. S. Dhillon, S. Hagan, O. Rath, and W. Kolch, MAP kinase signalling pathways in cancer, Oncogene, vol.26, issue.22, pp.3279-90, 2007.

R. Roskoski, ERK1/2 MAP kinases: structure, function, and regulation, Pharmacol Res, vol.66, issue.2, pp.105-148, 2012.

M. Maemura, Y. Iino, Y. Koibuchi, T. Yokoe, and Y. Morishita, Mitogen-activated protein kinase cascade in breast cancer, Oncology, vol.57, issue.2, pp.37-44, 1999.

J. Avruch, MAP kinase pathways: the first twenty years, Biochim Biophys Acta, vol.1773, issue.8, pp.1150-60, 2007.

T. Boutros, E. Chevet, and P. Metrakos, Mitogen-activated protein (MAP) kinase/MAP kinase phosphatase regulation: roles in cell growth, death, and cancer, Pharmacol Rev, vol.60, issue.3, pp.261-310, 2008.

K. Wennerberg, K. L. Rossman, and C. J. Der, The Ras superfamily at a glance, J Cell Sci, vol.118, pp.843-849, 2005.

C. A. Pratilas and D. B. Solit, Targeting the mitogen-activated protein kinase pathway: physiological feedback and drug response, Clin Cancer Res, vol.16, issue.13, pp.3329-3363, 2010.

M. Macaluso, G. Russo, C. Cinti, V. Bazan, N. Gebbia et al., Ras family genes: an interesting link between cell cycle and cancer, J Cell Physiol, vol.192, issue.2, pp.125-155, 2002.

E. Niemitz, Ras pathway activation in breast cancer, Nat Genet, vol.45, issue.11, p.1273, 2013.

F. C. Von-lintig, A. D. Dreilinger, N. M. Varki, A. M. Wallace, D. E. Casteel et al., Ras activation in human breast cancer, Breast Cancer Res Treat, vol.62, issue.1, pp.51-62, 2000.

C. Wellbrock, M. Karasarides, and R. Marais, The RAF proteins take centre stage, Nat Rev Mol Cell Biol, vol.5, issue.11, pp.875-85, 2004.

M. Beeram, A. Patnaik, and E. K. Rowinsky, Raf: a strategic target for therapeutic development against cancer, J Clin Oncol, vol.23, issue.27, pp.6771-90, 2005.

P. B. Chapman, A. Hauschild, C. Robert, J. B. Haanen, P. Ascierto et al., Improved survival with vemurafenib in melanoma with BRAF V600E mutation, N Engl J Med, vol.364, issue.26, pp.2507-2523, 2011.

A. Jylling, A. Rasmussen, E. Jakobsen, R. Christensen, and F. Sorensen, Are mutations in K-RAS, BRAF and PIK3CA genes critical for response to adjuvant trastuzumab treatment in patients with HER-2 positive breast cancer?, J Cancer Ther Res, vol.3, issue.1, 2014.

Y. D. Shaul and R. Seger, The MEK/ERK cascade: from signaling specificity to diverse functions, Biochim Biophys Acta, vol.1773, issue.8, pp.1213-1239, 2007.

J. M. Kyriakis and J. Avruch, Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update, Physiol Rev, vol.92, issue.2, pp.689-737, 2012.

J. W. Ramos, The regulation of extracellular signal-regulated kinase (ERK) in mammalian cells, Int J Biochem Cell Biol, vol.40, issue.12, pp.2707-2726, 2008.

J. A. Smith, C. E. Poteet-smith, Y. Xu, T. M. Errington, S. M. Hecht et al., Identification of the first specific inhibitor of p90 ribosomal S6 kinase (RSK) reveals an unexpected role for RSK in cancer cell proliferation, Cancer Res, vol.65, issue.3, pp.1027-1061, 2005.

H. G. Moon, J. K. Yi, H. S. Kim, H. Y. Lee, K. M. Lee et al., Phosphorylation of p90RSK is associated with increased response to neoadjuvant chemotherapy in ER-positive breast cancer, BMC Cancer, vol.12, p.585, 2012.

V. Serra, P. Eichhorn, G. Garc, and . Xed, RSK3/4 mediate resistance to PI3K pathway inhibitors in breast cancer, J Clin Invest, vol.123, issue.6, pp.2551-63, 2013.

M. S. Song, L. Salmena, and P. P. Pandolfi, The functions and regulation of the PTEN tumour suppressor, Nat Rev Mol Cell Biol, vol.13, issue.5, pp.283-96, 2012.

A. Cuenda and S. Rousseau, p38 MAP-kinases pathway regulation, function and role in human diseases, Biochim Biophys Acta, vol.1773, issue.8, pp.1358-75, 2007.

L. Chen, J. A. Mayer, T. I. Krisko, C. W. Speers, T. Wang et al., Inhibition of the p38 kinase suppresses the proliferation of human ER-negative breast cancer cells, Cancer Res, vol.69, issue.23, pp.8853-61, 2009.

J. L. Merlin, A. Harle, M. Lion, C. Ramacci, and A. Leroux, Expression and activation of P38 MAP kinase in invasive ductal breast cancers: correlation with expression of the estrogen receptor, HER2 and downstream signaling phosphorylated proteins, Oncol Rep, vol.30, issue.4, pp.1943-1951, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00866242

K. Ono and J. Han, The p38 signal transduction pathway: activation and function, Cell Signal, vol.12, issue.1, pp.1-13, 2000.

T. Zarubin and J. Han, Activation and signaling of the p38 MAP kinase pathway, Cell Res, vol.15, issue.1, pp.11-19, 2005.

L. New and J. Han, The p38 MAP kinase pathway and its biological function, Trends Cardiovasc Med, vol.8, issue.5, pp.220-228, 1998.

J. Whyte, O. Bergin, A. Bianchi, S. Mcnally, and F. Martin, Key signalling nodes in mammary gland development and cancer. Mitogen-activated protein kinase signalling in experimental models of breast cancer progression and in mammary gland development, Breast Cancer Res, vol.11, issue.5, p.209, 2009.

H. K. Koul, M. Pal, and S. Koul, Role of p38 MAP Kinase Signal Transduction in Solid Tumors, Genes Cancer, vol.4, issue.9, pp.342-59, 2013.

E. F. Wagner and A. R. Nebreda, Signal integration by JNK and p38 MAPK pathways in cancer development, Nat Rev Cancer, vol.9, issue.8, pp.537-586, 2009.

L. Tang and X. Han, The urokinase plasminogen activator system in breast cancer invasion and metastasis, Biomed Pharmacother, vol.67, issue.2, pp.179-82, 2013.

M. C. Gutierrez, S. Detre, S. Johnston, S. K. Mohsin, J. Shou et al., Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase, J Clin Oncol, vol.23, issue.11, pp.2469-76, 2005.

F. J. Esteva, A. A. Sahin, T. L. Smith, Y. Yang, L. Pusztai et al., Prognostic significance of phosphorylated P38 mitogen-activated protein kinase and HER-2 expression in lymph nodepositive breast carcinoma, Cancer, vol.100, issue.3, pp.499-506, 2004.

E. F. Wagner and A. R. Nebreda, Signal integration by JNK and p38 MAPK pathways in cancer development, Nat Rev Cancer, vol.9, issue.8, pp.537-586, 2009.

A. R. Green, D. G. Powe, E. A. Rakha, D. Soria, C. Lemetre et al., Identification of key clinical phenotypes of breast cancer using a reduced panel of protein biomarkers, Br J Cancer, vol.109, issue.7, pp.1886-94, 2013.

C. M. Perou, T. Sorlie, M. B. Eisen, M. Van-de-rijn, S. S. Jeffrey et al., Molecular portraits of human breast tumours, Nature, vol.406, issue.6797, pp.747-52, 2000.

T. Sorlie, C. M. Perou, R. Tibshirani, T. Aas, S. Geisler et al., Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications, Proc Natl Acad Sci, vol.98, issue.19, pp.10869-74, 2001.

T. Sorlie, R. Tibshirani, J. Parker, T. Hastie, J. S. Marron et al., Repeated observation of breast tumor subtypes in independent gene expression data sets, Proc Natl Acad Sci, vol.100, issue.14, pp.8418-8441, 2003.

C. Sotiriou, S. Y. Neo, L. M. Mcshane, E. L. Korn, P. M. Long et al., Breast cancer classification and prognosis based on gene expression profiles from a population-based study, Proc Natl Acad Sci, vol.100, issue.18, pp.10393-10401, 2003.

T. Sorlie, Y. Wang, C. Xiao, H. Johnsen, B. Naume et al., Distinct molecular mechanisms underlying clinically relevant subtypes of breast cancer: gene expression analyses across three different platforms, BMC Genomics, vol.7, p.127, 2006.

L. J. Van-'t-veer, H. Dai, M. J. Van-de-vijver, Y. D. He, A. A. Hart et al., Gene expression profiling predicts clinical outcome of breast cancer, Nature, vol.415, issue.6871, pp.530-536, 2002.

T. T. Hansel, H. Kropshofer, T. Singer, J. A. Mitchell, and A. J. George, The safety and side effects of monoclonal antibodies, Nat Rev Drug Discov, vol.9, issue.4, pp.325-363, 2010.

. Vidal, Résumé des caractéristiques du produit Herceptin®, 2013.

D. J. Slamon, B. Leyland-jones, S. Shak, H. Fuchs, V. Paton et al., Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2, N Engl J Med, vol.344, issue.11, pp.783-92, 2001.

J. J. Gemmete and S. K. Mukherji, Trastuzumab (Herceptin), AJNR Am J Neuroradiol, vol.32, issue.8, pp.1373-1377, 2011.

G. Valabrega, F. Montemurro, and M. Aglietta, Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer, Ann Oncol, vol.18, issue.6, pp.977-84, 2007.

J. Baselga, Treatment of HER2-overexpressing breast cancer, Ann Oncol, vol.21, issue.7, pp.36-40, 2010.

R. Nahta, D. Yu, M. C. Hung, G. N. Hortobagyi, and F. J. Esteva, Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer, Nat Clin Pract Oncol, vol.3, issue.5, pp.269-80, 2006.

M. Marty, F. Cognetti, D. Maraninchi, R. Snyder, L. Mauriac et al., Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group, J Clin Oncol, vol.23, pp.4265-74, 2005.

, HERCEPTIN® sous-cutanée: Avis de la Commission de la Transparence du 2 Avril, 2014.

S. R. Hubbard, EGF receptor inhibition: attacks on multiple fronts, Cancer Cell

S. Verma, D. Miles, L. Gianni, I. E. Krop, M. Welslau et al., Trastuzumab emtansine for HER2-positive advanced breast cancer, N Engl J Med, vol.367, pp.1783-91, 2012.

P. L. Mccormack, Pertuzumab: a review of its use for first-line combination treatment of HER2-positive metastatic breast cancer, Drugs, vol.73, issue.13, pp.1491-502, 2013.

J. Baselga, K. A. Gelmon, S. Verma, A. Wardley, P. Conte et al., Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy, J Clin Oncol, vol.28, issue.7, pp.1138-1182, 2010.

J. Baselga, J. Cortes, S. B. Kim, S. A. Im, R. Hegg et al., Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer, N Engl J Med, vol.366, issue.2, pp.109-128, 2012.

K. Miller, M. Wang, J. Gralow, M. Dickler, M. Cobleigh et al., Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer, N Engl J Med, vol.357, issue.26, pp.2666-76, 2007.

. Vidal, Résumé des caractéristiques du produit Avastin®, 2013.

. Avastin®, Avis de la Commission de la Transparence 25 mai, 2011.

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, vol.355, issue.26, pp.2733-2776, 2006.

B. Moy and P. E. Goss, Lapatinib: current status and future directions in breast cancer, Oncologist, vol.11, issue.10, pp.1047-57, 2006.

W. Xia, J. Bisi, J. Strum, L. Liu, K. Carrick et al., Regulation of survivin by ErbB2 signaling: therapeutic implications for ErbB2-overexpressing breast cancers, Cancer Res, vol.66, issue.3, pp.1640-1647, 2006.

. Vidal, Résumé des caractéristiques du produit TYVERB®, 2013.

M. Campone, P. Kerbrat, H. Roché, J. Bennouna, J. Cuillière et al., Prospects from the bench to the development of signal transduction inhibitors: application for breast cancer. Bull Cancer, 2003.

F. Andre, R. O'regan, M. Ozguroglu, M. Toi, B. Xu et al., Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial, Lancet Oncol, vol.15, issue.6, pp.580-91, 2014.

P. M. Lorusso, Mammalian target of rapamycin as a rational therapeutic target for breast cancer treatment, Oncology, vol.84, issue.1, pp.43-56, 2013.

E. B. Borders, C. Bivona, and P. J. Medina, Mammalian target of rapamycin: biological function and target for novel anticancer agents, Am J Health Syst Pharm, vol.67, issue.24, pp.2095-106, 2010.

H. Yang, D. G. Rudge, J. D. Koos, B. Vaidialingam, H. J. Yang et al., mTOR kinase structure, mechanism and regulation, Nature, vol.497, issue.7448, pp.217-240, 2013.

C. M. Barnett, Everolimus: targeted therapy on the horizon for the treatment of breast cancer, Pharmacotherapy, vol.32, issue.4, pp.383-96, 2012.

S. A. Hurvitz, F. Dalenc, M. Campone, R. M. O'regan, V. C. Tjan-heijnen et al., A phase 2 study of everolimus combined with trastuzumab and paclitaxel in patients with HER2-overexpressing advanced breast cancer that progressed during prior trastuzumab and taxane therapy, Breast Cancer Res Treat, vol.141, issue.3, pp.437-483, 2013.

, AFINITOR®: Avis de la Commission de la Transparence 3 Avril, HAS, 2013.

S. Oudard and R. T. Elaidi, Sequential therapy with targeted agents in patients with advanced renal cell carcinoma: optimizing patient benefit, Cancer Treat Rev, vol.38, issue.8, pp.981-988, 2012.

R. Nahta, Molecular Mechanisms of Trastuzumab-Based Treatment in HER2-Overexpressing Breast Cancer, ISRN Oncol, p.428062, 2012.

R. Nahta and F. J. Esteva, Trastuzumab: triumphs and tribulations, Oncogene, vol.26, issue.25, pp.3637-3680, 2007.

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.

P. J. Eichhorn, M. Gili, M. Scaltriti, V. Serra, M. Guzman et al., Phosphatidylinositol 3-kinase hyperactivation results in lapatinib resistance that is reversed by the mTOR/phosphatidylinositol 3-kinase inhibitor NVP-BEZ235, Cancer Res, vol.68, issue.22, pp.9221-9251, 2008.

Y. Samuels, L. A. Diaz, J. Schmidt-kittler, O. Cummins, J. M. Delong et al., Mutant PIK3CA promotes cell growth and invasion of human cancer cells, Cancer Cell, vol.7, issue.6, pp.561-73, 2005.

K. E. Bachman, P. Argani, Y. Samuels, N. Silliman, J. Ptak et al., The PIK3CA gene is mutated with high frequency in human breast cancers, Cancer Biol Ther, vol.3, issue.8, pp.772-777, 2004.

G. Perez-tenorio, L. Alkhori, B. Olsson, M. A. Waltersson, B. Nordenskjold et al., PIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancer, Clin Cancer Res, vol.13, issue.12, pp.3577-84, 2007.

A. Harle, M. Lion, N. Lozano, and J. L. Merlin, Clinical, diagnostic significance and theranostic interest of PIK3CA gene mutations in breast cancer

, Bull Cancer, vol.100, issue.10, pp.947-54, 2013.

B. Karakas, K. E. Bachman, and B. H. Park, Mutation of the PIK3CA oncogene in human cancers

, Br J Cancer, vol.94, issue.4, pp.455-464, 2006.

J. Dunlap, C. Le, A. Shukla, J. Patterson, A. Presnell et al., Phosphatidylinositol-3-kinase and AKT1 mutations occur early in breast carcinoma, Breast Cancer Res Treat, vol.120, issue.2, pp.409-427, 2010.

F. R. Mangone, I. G. Bobrovnitchaia, S. Salaorni, E. Manuli, and M. A. Nagai, PIK3CA exon 20 mutations are associated with poor prognosis in breast cancer patients, Clinics, vol.67, issue.11, pp.1285-90, 2012.

K. Kalinsky, L. M. Jacks, A. Heguy, S. Patil, M. Drobnjak et al., PIK3CA mutation associates with improved outcome in breast cancer, Clin Cancer Res, vol.15, issue.16, pp.5049-59, 2009.

F. L. Chen, W. Xia, and N. L. Spector, Acquired resistance to small molecule ErbB2 tyrosine kinase inhibitors, Clin Cancer Res, vol.14, issue.21, pp.6730-6734, 2008.

S. Guo and G. E. Sonenshein, Forkhead box transcription factor FOXO3a regulates estrogen receptor alpha expression and is repressed by the Her-2/neu/phosphatidylinositol 3-kinase/Akt signaling pathway, Mol Cell Biol, vol.24, pp.8681-90, 2004.

T. Trowe, S. Boukouvala, K. Calkins, R. E. Cutler, J. 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.

J. W. Lee, Y. H. Soung, S. H. Seo, S. Y. Kim, C. H. Park et al., Somatic mutations of ERBB2 kinase domain in gastric, colorectal, and breast carcinomas, Clin Cancer Res, vol.12, issue.1, pp.57-61, 2006.

E. Luporsi, Validation des biomarqueurs, niveau de preuve et essais cliniques adaptatifs. Les biomarqueurs moléculaires en oncologie, sous la direction de Jean-Louis Merlin, pp.19-27, 2014.

M. Cizkova, M. E. Dujaric, J. Lehmann-che, V. Scott, O. Tembo et al., Outcome impact of PIK3CA mutations in HER2-positive breast cancer patients treated with trastuzumab, Br J Cancer, vol.108, issue.9, pp.1807-1816, 2013.

F. J. Esteva, H. Guo, S. Zhang, C. Santa-maria, S. Stone et al., status: association with trastuzumab response and survival in patients with HER2-positive metastatic breast cancer, Am J Pathol, vol.3, issue.4, pp.1647-56, 2010.

J. D. Jensen, A. Knoop, A. V. Laenkholm, M. Grauslund, M. B. Jensen et al., PIK3CA mutations, PTEN, and pHER2 expression and impact on outcome in HER2-positive early-stage breast cancer patients treated with adjuvant chemotherapy and trastuzumab

, Ann Oncol, vol.23, issue.8, pp.2034-2076, 2012.

V. Abramson and C. L. Arteaga, New strategies in HER2-overexpressing breast cancer: many combinations of targeted drugs available, Clin Cancer Res, vol.17, issue.5, pp.952-960, 2011.

L. Orlando, P. Schiavone, P. Fedele, N. Calvani, A. Nacci et al., Molecularly targeted endocrine therapies for breast cancer, Cancer Treat Rev, vol.36, issue.3, pp.67-71, 2010.

T. W. Miller, J. M. Balko, and C. L. Arteaga, Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer, J Clin Oncol, vol.29, issue.33, pp.4452-61, 2011.

C. X. Ma, R. J. Crowder, and M. J. Ellis, Importance of PI3-kinase pathway in response/resistance to aromatase inhibitors, Steroids, vol.76, issue.8, pp.750-752, 2011.

S. Loi, S. Michiels, J. Baselga, J. M. Bartlett, S. K. Singhal et al., PIK3CA genotype and a PIK3CA mutation-related gene signature and response to everolimus and letrozole in estrogen receptor positive breast cancer, PLoS One, vol.8, issue.1, p.53292, 2013.

S. M. Donnelly, E. Paplomata, B. M. Peake, E. Sanabria, Z. Chen et al., P38 MAPK contributes to resistance and invasiveness of HER2-overexpressing breast cancer, Curr Med Chem, vol.21, issue.4, pp.501-511, 2014.

C. H. Lu, S. L. Wyszomierski, L. M. Tseng, M. H. Sun, K. H. Lan et al., Preclinical testing of clinically applicable strategies for overcoming trastuzumab resistance caused by PTEN deficiency, Clin Cancer Res, vol.13, issue.19, pp.5883-5891, 2007.

P. K. Morrow, G. M. Wulf, J. Ensor, D. J. Booser, J. A. Moore et al., Phase I/II study of trastuzumab in combination with everolimus (RAD001) in patients with HER2-overexpressing metastatic breast cancer who progressed on trastuzumab-based therapy, J Clin Oncol, vol.29, issue.23, pp.3126-3158, 2011.

F. Andre, M. Campone, R. O'regan, C. Manlius, C. Massacesi et al., Phase I study of everolimus plus weekly paclitaxel and trastuzumab in patients with metastatic breast cancer pretreated with trastuzumab, J Clin Oncol, vol.28, issue.34, pp.5110-5115, 2010.

S. A. Hurvitz, F. Dalenc, M. Campone, R. M. O'regan, V. C. Tjan-heijnen et al., A phase 2 study of everolimus combined with trastuzumab and paclitaxel in patients with HER2-overexpressing advanced breast cancer that progressed during prior trastuzumab and taxane therapy, Breast Cancer Res Treat, vol.141, issue.3, pp.437-483, 2013.

S. A. Hurvitz, F. Andre, Z. Jiang, Z. Shao, M. S. Mano et al., Combination of everolimus with trastuzumab plus paclitaxel as first-line treatment for patients with HER2-positive advanced breast cancer (BOLERO-1): a phase 3, randomised, double-blind, multicentre trial, Lancet Oncol, vol.16, issue.7, pp.816-845, 2015.

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, pp.1673-84, 2005.

M. J. Piccart-gebhart, M. Procter, B. Leyland-jones, A. Goldhirsch, M. Untch et al., Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer, N Engl J Med, vol.353, issue.16, pp.1659-72, 2005.

R. Gennari, S. Menard, F. Fagnoni, L. Ponchio, M. Scelsi et al., Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2, Clin Cancer Res, vol.10, issue.17, pp.5650-5655, 2004.

B. P. Coudert, R. Largillier, L. Arnould, P. Chollet, M. Campone et al., Multicenter phase II trial of neoadjuvant therapy with trastuzumab, docetaxel, and carboplatin for human epidermal growth factor receptor-2-overexpressing stage II or III breast cancer: results of the GETN(A)-1 trial, J Clin Oncol, vol.25, pp.2678-84, 2007.

B. P. Coudert, L. Arnould, L. Moreau, P. Chollet, B. Weber et al., Preoperative systemic (neo-adjuvant) therapy with trastuzumab and docetaxel for HER2-overexpressing stage II or III breast cancer: results of a multicenter phase II trial, Ann Oncol, vol.17, issue.3, pp.409-423, 2006.

A. U. Buzdar, N. K. Ibrahim, D. Francis, D. J. Booser, E. S. Thomas et al., Significantly higher pathologic complete remission rate after neoadjuvant therapy with trastuzumab, paclitaxel, and epirubicin chemotherapy: results of a randomized trial in human epidermal growth factor receptor 2-positive operable breast cancer, J Clin Oncol, vol.23, issue.16, pp.3676-85, 2005.

F. Chergui, A. S. Chretien, S. Bouali, C. Ramacci, M. Rouyer et al., Validation of a phosphoprotein array assay for characterization of human tyrosine kinase receptor downstream signaling in breast cancer, Clin Chem, vol.55, issue.7, pp.1327-1363, 2009.

C. Denkert, S. Darb-esfahani, S. Loibl, I. Anagnostopoulos, and K. Johrens, Anti-cancer immune response mechanisms in neoadjuvant and targeted therapy, Semin Immunopathol, vol.33, issue.4, pp.341-51, 2011.

R. A. Clynes, T. L. Towers, L. G. Presta, and J. V. Ravetch, Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets, Nat Med, vol.6, issue.4, pp.443-449, 2000.

H. E. Kohrt, R. Houot, K. Weiskopf, M. J. Goldstein, F. Scheeren et al., Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer, J Clin Invest, vol.122, issue.3, pp.1066-75, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00869052

S. Loi, Tumor-infiltrating lymphocytes, breast cancer subtypes and therapeutic efficacy, Oncoimmunology, vol.2, issue.7, p.24720, 2013.

S. Loi, S. Michiels, R. Salgado, N. Sirtaine, V. Jose et al., Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial, Ann Oncol, vol.25, issue.8, pp.1544-50, 2014.

F. Andre, M. V. Dieci, P. Dubsky, C. Sotiriou, G. Curigliano et al., Molecular pathways: involvement of immune pathways in the therapeutic response and outcome in breast cancer, Clin Cancer Res, vol.19, issue.1, pp.28-33, 2013.

A. Beano, E. Signorino, A. Evangelista, D. Brusa, M. Mistrangelo et al., Correlation between NK function and response to trastuzumab in metastatic breast cancer patients, J Transl Med, vol.6, p.25, 2008.

M. Untch, M. Rezai, S. Loibl, P. A. Fasching, J. Huober et al., Neoadjuvant treatment with trastuzumab in HER2-positive breast cancer: results from the GeparQuattro study, J Clin Oncol, vol.28, issue.12, pp.2024-2055, 2010.

E. Tagliabue, M. Campiglio, S. M. Pupa, S. Menard, and A. Balsari, Activity and resistance of trastuzumab according to different clinical settings, Cancer Treat Rev, vol.38, issue.3, pp.212-219, 2012.

A. Carracedo and P. P. Pandolfi, The PTEN-PI3K pathway: of feedbacks and cross-talks, Oncogene, vol.27, issue.41, pp.5527-5568, 2008.

V. Serra, M. Scaltriti, L. Prudkin, P. J. Eichhorn, Y. H. Ibrahim et al., PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer, Oncogene, vol.30, issue.22, pp.2547-57, 2011.

H. P. Soares, Y. Ni, K. Kisfalvi, J. Sinnett-smith, and E. Rozengurt, Different patterns of Akt and ERK feedback activation in response to rapamycin, active-site mTOR inhibitors and metformin in pancreatic cancer cells, PLoS One, vol.8, issue.2, p.57289, 2013.

L. Arnould, M. Gelly, F. Penault-llorca, L. Benoit, F. Bonnetain et al., Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism?, Br J Cancer, vol.94, issue.2, pp.259-67, 2006.

S. K. Mohsin, H. L. Weiss, M. C. Gutierrez, G. C. Chamness, R. Schiff et al., Neoadjuvant trastuzumab induces apoptosis in primary breast cancers, J Clin Oncol, vol.23, issue.11, pp.2460-2468, 2005.

R. E. Board, N. J. Thelwell, P. F. Ravetto, S. Little, M. Ranson et al., Multiplexed assays for detection of mutations in PIK3CA, Clin Chem, vol.54, issue.4, pp.757-60, 2008.

V. Hofman, M. Ilie, V. Gavric-tanga, V. Lespinet, M. Mari et al.,

, Ann Pathol, vol.30, issue.2, pp.85-93, 2010.

I. F. Pinhel, F. A. Macneill, M. J. Hills, J. Salter, S. Detre et al., Extreme loss of immunoreactive p-Akt and p-Erk1/2 during routine fixation of primary breast cancer, Breast Cancer Res, vol.12, issue.5, p.76, 2010.

S. Siddiqui and D. L. Rimm, Pre-analytic variables and phospho-specific antibodies: the Achilles heel of immunohistochemistry, Breast Cancer Res, vol.12, issue.6, p.113, 2010.

K. David and H. Juhl, Immunohistochemical Detection of Phosphoproteins and Cancer Pathways, Handbook of Practical Immunohistochemistry, pp.85-90, 2015.

E. A. Trigka, G. Levidou, A. A. Saetta, I. Chatziandreou, P. Tomos et al., A detailed immunohistochemical analysis of the PI3K/AKT/mTOR pathway in lung cancer: correlation with PIK3CA, AKT1, K-RAS or PTEN mutational status and clinicopathological features, Oncol Rep, vol.30, issue.2, pp.623-659, 2013.

G. Hudelist, W. J. Kostler, K. Czerwenka, E. Kubista, J. Attems et al., Her-2/neu and EGFR tyrosine kinase activation predict the efficacy of trastuzumab-based therapy in patients with metastatic breast cancer, Int J Cancer, vol.118, issue.5, pp.1126-1160, 2006.

E. Felip, F. Rojo, M. Reck, A. Heller, B. Klughammer et al., A phase II pharmacodynamic study of erlotinib in patients with advanced non-small cell lung cancer previously treated with platinum-based chemotherapy, Clin Cancer Res, vol.14, issue.12, pp.3867-74, 2008.

J. Merlin, L. M. Wong, J. Bachelot, T. Andre, F. Treilleux et al., Abstract P3-06-07: Alterations of intratumoral signalling in breast cancer patients receiving pre-operative trastuzumab alone or combined with everolimus, Cancer Research, vol.75, issue.9, pp.3-06, 2015.

S. Zimmermann and K. Moelling, Phosphorylation and regulation of Raf by Akt (protein kinase B), Science, vol.286, issue.5445, pp.1741-1745, 1999.

K. L. Guan, C. Figueroa, T. R. Brtva, T. Zhu, J. Taylor et al., Negative regulation of the serine/threonine kinase B-Raf by Akt, J Biol Chem, vol.275, issue.35, pp.27354-27363, 2000.

Z. Q. Yuan, R. I. Feldman, G. E. Sussman, D. Coppola, S. V. Nicosia et al., AKT2 inhibition of cisplatin-induced JNK/p38 and Bax activation by phosphorylation of ASK1: implication of AKT2 in chemoresistance, J Biol Chem, vol.278, issue.26, pp.23432-23472, 2003.

I. Vivanco, N. Palaskas, C. Tran, S. P. Finn, G. Getz et al., Identification of the JNK signaling pathway as a functional target of the tumor suppressor PTEN, Cancer Cell, vol.11, issue.6, pp.555-69, 2007.

J. Gu, M. Tamura, and K. M. Yamada, Tumor suppressor PTEN inhibits integrin-and growth factor-mediated mitogen-activated protein (MAP) kinase signaling pathways, J Cell Biol, vol.143, issue.5, pp.1375-83, 1998.

P. P. Roux and J. Blenis, ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions, Microbiol Mol Biol Rev, vol.68, issue.2, pp.320-364, 2004.

L. Ma, Z. Chen, H. Erdjument-bromage, P. Tempst, and P. P. Pandolfi, Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis, Cell, vol.121, issue.2, pp.179-93, 2005.

G. Finak, N. Bertos, F. Pepin, S. Sadekova, M. Souleimanova et al., Stromal gene expression predicts clinical outcome in breast cancer, Nat Med, vol.14, issue.5, pp.518-545, 2008.

Y. Mao, E. T. Keller, D. H. Garfield, K. Shen, and J. Wang, Stromal cells in tumor microenvironment and breast cancer, Cancer Metastasis Rev, vol.32, issue.1-2, pp.303-318, 2013.

S. Winslow, K. Leandersson, A. Edsjö, and C. Larsson, Prognostic stromal gene signatures in breast cancer, Breast Cancer Res, vol.17, issue.1, p.23, 2015.

E. M. De-kruijf, J. G. Van-nes, C. J. Van-de-velde, H. Putter, V. T. Smit et al., Tumorstroma ratio in the primary tumor is a prognostic factor in early breast cancer patients, especially in triple-negative carcinoma patients, Breast Cancer Res Treat, vol.125, issue.3, pp.687-96, 2011.

C. L. Downey, H. H. Thygesen, N. Sharma, and A. M. Shaaban, Prognostic significance of tumour stroma ratio in inflammatory breast cancer, Springerplus, vol.4, p.68, 2015.

E. V. Denisov, N. V. Litviakov, M. V. Zavyalova, V. M. Perelmuter, S. V. Vtorushin et al., Intratumoral morphological heterogeneity of breast cancer: neoadjuvant chemotherapy efficiency and multidrug resistance gene expression, Sci Rep, vol.4, p.4709, 2014.

P. Farmer, H. Bonnefoi, P. Anderle, D. Cameron, P. Wirapati et al., A stromarelated gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer, Nat Med, vol.15, issue.1, pp.68-74, 2009.

M. Scartozzi, I. Bearzi, R. Berardi, A. Mandolesi, C. Pierantoni et al., Epidermal growth factor receptor (EGFR) downstream signalling pathway in primary colorectal tumours and related metastatic sites: optimising EGFR-targeted treatment options, Br J Cancer, vol.97, issue.1, pp.92-99, 2007.

M. Murtaza, S. J. Dawson, D. W. Tsui, D. Gale, T. Forshew et al., Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA, Nature, vol.497, issue.7447, pp.108-120, 2013.

P. C. Nowell, The clonal evolution of tumor cell populations, Science, vol.194, issue.4260, pp.23-31, 1976.

M. Greaves and C. C. Maley, Clonal evolution in cancer, Nature, vol.481, issue.7381, pp.306-319, 2012.

C. K. Ng, L. G. Martelotto, A. Gauthier, H. C. Wen, S. Piscuoglio et al., Intra-tumor genetic heterogeneity and alternative driver genetic alterations in breast cancers with heterogeneous HER2 gene amplification, Genome Biol, vol.16, p.107, 2015.

M. Shackleton, E. Quintana, E. R. Fearon, and S. J. Morrison, Heterogeneity in cancer: cancer stem cells versus clonal evolution, Cell, vol.138, issue.5, pp.822-831, 2009.

M. Gerlinger, A. J. Rowan, S. Horswell, J. Larkin, D. Endesfelder et al., Intratumor heterogeneity and branched evolution revealed by multiregion sequencing, N Engl J Med, vol.366, issue.10, pp.883-92, 2012.

F. Mouliere and A. R. Thierry, The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients, Expert Opin Biol Ther, vol.12, issue.1, pp.209-224, 2012.

E. Gormally, E. Caboux, P. Vineis, and P. Hainaut, Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance, Mutat Res, vol.635, issue.2-3, pp.105-122, 2007.

J. L. Ramirez, M. Taron, C. Balana, C. Sarries, P. Mendez et al., Serum DNA as a tool for cancer patient management, Rocz Akad Med Bialymst, vol.48, pp.34-41, 2003.

J. Madic, S. Piperno-neumann, V. Servois, A. Rampanou, M. Milder et al., Pyrophosphorolysis-activated polymerization detects circulating tumor DNA in metastatic uveal melanoma, Clin Cancer Res, vol.18, issue.14, pp.3934-3975, 2012.

S. J. Dawson, D. W. Tsui, M. Murtaza, H. Biggs, O. M. Rueda et al., Analysis of circulating tumor DNA to monitor metastatic breast cancer, N Engl J Med, vol.368, issue.13, pp.1199-209, 2013.

E. Crowley, D. Nicolantonio, F. Loupakis, F. Bardelli, and A. , Liquid biopsy: monitoring cancergenetics in the blood, Nat Rev Clin Oncol, vol.10, issue.8, pp.472-84, 2013.

A. L. Richardson and J. D. Iglehart, BEAMing up personalized medicine: mutation detection in blood, Clin Cancer Res, vol.18, issue.12, pp.3209-3220, 2012.

R. Le, Les progrès de la biologie moléculaire ont permis la caractérisation des principales voies de signalisation et ont mis en évidence l'implication majeure de la signalisation cellulaire dans les processus de cancérogenèse. Des cibles moléculaires ont ainsi été identifiées et ont permis le développement de thérapeutiques dites ciblées, telles que les anticorps monoclonaux ou encore les inhibiteurs de kinase. Malgré ces avancées considérables qui ont permis l'amélioration de la prise en charge des patientes, on constate l'apparition de résistances aux traitements. Ce travail avait pour objectifs d'identifier de nouveaux biomarqueurs et de déterminer leur signification clinique

, Dans un premier temps nous avons étudié les mutations activatrices du gène PIK3CA. Ces mutations sont retrouvées dans 25% des cancers du sein et sont impliquées dans la résistance au trastuzumab, aux anti-oestrogènes et aux inhibiteurs de mTOR

, Les résultats des 2 techniques étaient concordants (?=0,845 ; p<0,001) et une relation entre mutations du gène PIK3CA et grade SBR a été mise en évidence, les tumeurs de grade SBR III étant moins fréquemment mutées que les autres, échantillons de tumeurs de sein infiltrantes ont été analysés par une technique de PCR-HRM

/. Akt/mtor and R. Et-p38mapkinase, /2 (p=0.003) et p-P38MAPK (p<0.001) dans le bras de traitement associant l'évérolimus au trastuzumab qui pourrait s'expliquer par la suppression par l'évérolimus d'une boucle de rétrocontrôle négatif contrôlant l'activation de la voie RAS/RAF/MAPKinases. Dans le bras de traitement évaluant le trastuzumab seul, aucune variation du niveau d'expression des phosphoprotéines n'a été mise en évidence, y compris en aval du récepteur HER2, ce qui soulève l'hypothèse d'un mécanisme d'action prédominant immunologique du trastuzumab, Pour cela nous avons analysé le niveau d'expression des phosphoprotéines p-AKT, p-GSK3?, p-S6 kinase, vol.2, p.1

. Dans-son-ensemble, Advances in molecular biology have allowed the characterization of the major signaling pathways and revealed their major implication in carcinogenesis processes. Molecular targets have been identified and have enabled the development of targeted therapies, such as monoclonal antibodies, or kinase inhibitors. Despite these considerable advances that have improved the care of patients, emerging of resistance to treatments has been observed. The aim of this work was to identify new tumor biomarkers and determine their clinical significance, ce travail a mis en évidence que la détermination du statut mutationnel du gène PIK3CA et du niveau d'

, We analyzed 149 invasive breast tumor samples for PIK3CA gene mutations by PCR-HRM (High Resolution Melting) and 118 by PCR-ARMS (Amplification Refractory Mutation System). The results achieved with the 2 techniques were consistent (? = 0.845; p <0.001) and a relationship between PIK3CA mutations and grade SBR was highlighted with a lower occurrence of mutations in SBR grade III tumors

. Secondly, This part includes 3 studies. The first study was a retrospective study of 45 frozen samples of invasive breast tumors in which we observed that the level of expression of P38 and p-P38 was higher in the ER + tumors. The second study was a prospective study to identify biomarkers of response to trastuzumabeverolimus association in patients with early breast cancer treated preoperatively. This study showed a statistically significant increase of the expression level of p-MEK1 (p = 0.012), p-ERK1/2 (p = 0.003) and p-p38MAPK (p<0.001) in arm treated by trastuzumab associated with everolimus. It could be explained by the repression of a negative feedback loop involving S6K, PI3K and RAS by everolimus, leading to the activation of RAS/RAF/MAPKinases signaling pathway. In the control arm investigating trastuzumab alone, no significant variations of the level of expression of phosphoproteins was demonstrated, raising the hypothesis of the implementation of a predominant immunological mechanism of action for Trastuzumab. The third study that compared effect of trastuzumab in vitro, RAS/RAF/MAPKinases and P38MAPKinase signaling pathways. We have analyzed the expression level of phosphoproteins p-AKT, p-GSK3?, p-S6 kinase, p.90

, As a whole, this work showed that determining the mutation status of PIK3CA and the expression level of phosphoproteins could be useful to refine the molecular characterization of breast cancers and optimize the criteria used to personalize the prescription of targeted therapies