A. Stuckey, Breast Cancer, Clinical Obstetrics and Gynecology, vol.54, issue.1, pp.96-102, 2011.
DOI : 10.1097/GRF.0b013e3182080056

A. Goldhirsch, J. H. Glick, R. D. Gelher, and H. J. Senn, Meeting Highlights: International Consensus Panel on the Treatment of Primary Breast Cancer, JNCI Journal of the National Cancer Institute, vol.90, issue.21, pp.1601-1608, 1998.
DOI : 10.1093/jnci/90.21.1601

C. Early-breast, . Trialists-'collaborative, and . Group, Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials, The Lancet, vol.365, issue.9472, pp.1687-1717, 2005.

M. De-laurentiis, G. Cancello, and D. Agostino, Taxane-Based Combinations As Adjuvant Chemotherapy of Early Breast Cancer: A Meta-Analysis of Randomized Trials, Journal of Clinical Oncology, vol.26, issue.1, pp.44-53, 2008.
DOI : 10.1200/JCO.2007.11.3787

E. H. Romond, E. A. Perez, and J. Bryant, Trastuzumab plus Adjuvant Chemotherapy for Operable HER2-Positive Breast Cancer, New England Journal of Medicine, vol.353, issue.16, pp.1673-1684, 2005.
DOI : 10.1056/NEJMoa052122

J. Cuzick, I. Sestak, and S. E. Pinder, Effect of tamoxifen and radiotherapy in women with locally excised ductal carcinoma in situ: long-term results from the UK/ANZ DCIS trial, The Lancet Oncology, vol.12, issue.1, pp.21-29, 2011.
DOI : 10.1016/S1470-2045(10)70266-7

D. W. Miles, A. Chan, and L. Y. Dirix, Phase III Study of Bevacizumab Plus Docetaxel Compared With Placebo Plus Docetaxel for the First-Line Treatment of Human Epidermal Growth Factor Receptor 2???Negative Metastatic Breast Cancer, Journal of Clinical Oncology, vol.28, issue.20, pp.3239-3247, 2010.
DOI : 10.1200/JCO.2008.21.6457

URL : https://hal.archives-ouvertes.fr/inserm-00508083

M. Andersson, E. Lidbrink, and K. Bjerre, Phase III Randomized Study Comparing Docetaxel Plus Trastuzumab With Vinorelbine Plus Trastuzumab As First-Line Therapy of Metastatic or Locally Advanced Human Epidermal Growth Factor Receptor 2???Positive Breast Cancer: The HERNATA Study, Journal of Clinical Oncology, vol.29, issue.3, pp.264-271, 2011.
DOI : 10.1200/JCO.2010.30.8213

J. F. Robertson, A. Llombart-cussac, and J. Rolski, Activity of Fulvestrant 500 mg Versus Anastrozole 1 mg As First-Line Treatment for Advanced Breast Cancer: Results From the FIRST Study, Journal of Clinical Oncology, vol.27, issue.27, pp.4530-4535, 2009.
DOI : 10.1200/JCO.2008.21.1136

M. Lacroix, R. A. Toillon, and G. Leclercq, p53 and breast cancer, an update, Endocrine Related Cancer, vol.13, issue.2, pp.293-325, 2006.
DOI : 10.1677/erc.1.01172

B. Vogelstein, A deadly inheritance, Nature, vol.348, issue.6303, pp.681-682, 1990.
DOI : 10.1038/348681a0

T. Soussi, C. Caron-de-fromentel, and P. May, Structural aspects of the p53 protein in relation to gene evolution, Oncogene, vol.5, issue.7, pp.945-952, 1990.

E. A. Slee, D. J. O-'connor, and X. Lu, To die or not to die: how does p53 decide?, Oncogene, vol.23, issue.16, pp.2809-2818, 2004.
DOI : 10.1038/sj.onc.1207516

T. Soussi, The p53 Tumor Suppressor Gene: From Molecular Biology to Clinical Investigation, Annals of the New York Academy of Sciences, vol.17, issue.1, pp.121-137, 2000.
DOI : 10.1111/j.1749-6632.2000.tb06705.x

M. V. Poyurovsky, C. Katz, and O. Laptenko, The C terminus of p53 binds the N-terminal domain of MDM2, Nature Structural & Molecular Biology, vol.3, issue.8, pp.982-989, 2010.
DOI : 10.1101/gad.1795709

M. A. Lohrum, D. B. Woods, R. L. Ludwig, E. Bálint, and K. H. Vousden, C-Terminal Ubiquitination of p53 Contributes to Nuclear Export, Molecular and Cellular Biology, vol.21, issue.24, pp.8521-8532, 2001.
DOI : 10.1128/MCB.21.24.8521-8532.2001

G. He, Z. H. Siddik, and Z. Huang, Induction of p21 by p53 following DNA damage inhibits both Cdk4 and Cdk2 activities, Oncogene, vol.21, issue.18, pp.2929-2943, 2005.
DOI : 10.1038/sj.onc.1208474

M. L. Cox and D. W. Meek, Phosphorylation of serine 392 in p53 is a common and integral event during p53 induction by diverse stimuli, Cellular Signalling, vol.22, issue.3, pp.564-571, 2010.
DOI : 10.1016/j.cellsig.2009.11.014

L. Wiesmüller, Genetic Stabilization by p53 Involves Growth Regulatory and Repair Pathways, Journal of Biomedicine and Biotechnology, vol.1, issue.1, pp.7-10, 2001.
DOI : 10.1155/S1110724301000043

M. Ocker and R. Schneider-stock, Histone deacetylase inhibitors: Signalling towards p21cip1/waf1, The International Journal of Biochemistry & Cell Biology, vol.39, issue.7-8, pp.1367-1374, 2007.
DOI : 10.1016/j.biocel.2007.03.001

E. S. Helton and X. Chen, p53 modulation of the DNA damage response, Journal of Cellular Biochemistry, vol.25, issue.4, pp.883-896, 2007.
DOI : 10.1002/jcb.21091

I. Ben-porath and R. A. Weinberg, The signals and pathways activating cellular senescence, The International Journal of Biochemistry & Cell Biology, vol.37, issue.5, pp.961-976, 2005.
DOI : 10.1016/j.biocel.2004.10.013

G. M. Wahl and A. M. Carr, The evolution of diverse biological responses to DNA damage: insights from yeast and p53, Nature Cell Biology, vol.3, issue.12, pp.277-286, 2001.
DOI : 10.1038/ncb1201-e277

K. M. Ryan, p53 and autophagy in cancer: Guardian of the genome meets guardian of the proteome, European Journal of Cancer, vol.47, issue.1, pp.44-50, 2011.
DOI : 10.1016/j.ejca.2010.10.020

M. Mihara, S. Erster, and A. Zaika, p53 Has a Direct Apoptogenic Role at the Mitochondria, Molecular Cell, vol.11, issue.3, pp.577-590, 2003.
DOI : 10.1016/S1097-2765(03)00050-9

F. Rodier, J. Campisi, and D. Bhaumik, Two faces of p53: aging and tumor suppression, Nucleic Acids Research, vol.35, issue.22, pp.7475-7484, 2007.
DOI : 10.1093/nar/gkm744

M. C. Maiuri, L. Galluzzi, E. Morselli, O. Kepp, S. A. Malik et al., Autophagy regulation by p53, Current Opinion in Cell Biology, vol.22, issue.2, pp.181-185, 2010.
DOI : 10.1016/j.ceb.2009.12.001

A. V. Budanov and M. Karin, p53 Target Genes Sestrin1 and Sestrin2 Connect Genotoxic Stress and mTOR Signaling, Cell, vol.134, issue.3, pp.451-460, 2008.
DOI : 10.1016/j.cell.2008.06.028

D. Crighton, S. Wilkinson, and J. O. Prey, DRAM, a p53-Induced Modulator of Autophagy, Is Critical for Apoptosis, Cell, vol.126, issue.1, pp.121-134, 2006.
DOI : 10.1016/j.cell.2006.05.034

E. Tasdemir, M. C. Maiuri, and L. Galluzzi, Regulation of autophagy by cytoplasmic p53, Nature Cell Biology, vol.13, issue.6, pp.676-687, 2008.
DOI : 10.1093/emboj/19.21.5720

E. Morselli, E. Tasdemir, and M. C. Maiuri, Mutant p53 protein localized in the cytoplasm inhibits autophagy, Cell Cycle, vol.7, issue.19, pp.3056-3061, 2008.
DOI : 10.4161/cc.7.19.6751

H. Vakifahmetoglu, M. Olsson, and B. Zhivotovsky, Death through a tragedy: mitotic catastrophe, Cell Death and Differentiation, vol.5, issue.7, pp.1153-1162, 2008.
DOI : 10.1002/(SICI)1097-0185(199605)245:1<1::AID-AR1>3.0.CO;2-2

M. Castedo, J. L. Perfettini, T. Roumier, K. Andreau, R. Medema et al., Cell death by mitotic catastrophe: a molecular definition, Oncogene, vol.23, issue.16, pp.2825-2837, 2004.
DOI : 10.1038/sj.onc.1207528

S. L. Harris and A. J. Levine, The p53 pathway: positive and negative feedback loops, Oncogene, vol.12, issue.17, pp.2899-2908, 2005.
DOI : 10.1038/sj.onc.1208615

J. G. Teodoro, S. K. Evans, and M. R. Green, Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome, p53, vessel count, and vascular endothelial growth factor expression in human colon cancer, pp.1175-1186, 1998.
DOI : 10.1007/s00109-007-0221-2

P. Faviana, L. Boldrini, and R. Spisni, Neoangiogenesis in colon cancer: Correlation between vascular density, vascular endothelial growth factor (VEGF) and p53 protein expression, Oncology Reports, vol.9, issue.3, pp.617-620, 2002.
DOI : 10.3892/or.9.3.617

P. H. Maxwell, C. W. Pugh, and P. J. Ratcliffe, Activation of the HIF pathway in cancer, Current Opinion in Genetics & Development, vol.11, issue.3, pp.293-299, 2001.
DOI : 10.1016/S0959-437X(00)00193-3

X. Q. Wang, E. J. Stanbridge, X. Lao, Q. Cai, S. T. Fan et al., Arrest, Radiation Research, vol.168, issue.6, pp.706-715, 2007.
DOI : 10.1667/RR1075.1

URL : https://hal.archives-ouvertes.fr/hal-00719604

T. Li, R. Santockyte, and R. F. Shen, Expression of SUMO-2/3 Induced Senescence through p53- and pRB-mediated Pathways, Journal of Biological Chemistry, vol.281, issue.47, pp.36221-36227, 2006.
DOI : 10.1074/jbc.M608236200

L. E. Giono and J. J. Manfredi, Mdm2 Plays a Positive Role as an Effector of p53-Dependent Responses, Cell Cycle, vol.6, issue.17, pp.2143-2147, 2007.
DOI : 10.4161/cc.6.17.4647

F. Toledo and G. M. Wahl, Regulating the p53 pathway: in vitro hypotheses, in vivo veritas, Nature Reviews Cancer, vol.55, issue.12, pp.909-923, 2006.
DOI : 10.1002/gcc.20310

S. Geisler, P. E. Lonning, and T. Aas, Influence of TP53 gene alterations and c-erbB-2 expression on the response to treatment with doxorubicin in locally advanced breast cancer, Cancer Research, vol.61, issue.6, pp.2505-2512, 2001.

F. C. Schmitt, R. Soares, L. Cirnes, and R. Seruca, P53 in Breast Carcinomas: Association Between Presence of Mutation and Immunohistochemical Expression Using a Semiquantitative Approach, Pathology - Research and Practice, vol.194, issue.12, pp.815-819, 1998.
DOI : 10.1016/S0344-0338(98)80083-6

J. M. Flaman, T. Frebourg, and V. Moreau, A simple p53 functional assay for screening cell lines, blood, and tumors., Proceedings of the National Academy of Sciences, vol.92, issue.9, pp.3963-3967, 1995.
DOI : 10.1073/pnas.92.9.3963

P. Bertheau, M. Espié, and E. Turpin, <i>TP53</i> Status and Response to Chemotherapy in Breast Cancer, Pathobiology, vol.75, issue.2, pp.132-139, 2008.
DOI : 10.1159/000123851

M. Varna, H. Soliman, and J. P. Feugeas, Changes in allelic imbalances in locally advanced breast cancers after chemotherapy, British Journal of Cancer, vol.18, issue.8, pp.1157-1164, 2007.
DOI : 10.1038/sj.onc.1200812

E. Manie, A. Vincent-salomon, and J. Lehmann-che, High Frequency of TP53 Mutation in BRCA1 and Sporadic Basal-like Carcinomas but not in BRCA1 Luminal Breast Tumors, Cancer Research, vol.69, issue.2, pp.663-671, 2009.
DOI : 10.1158/0008-5472.CAN-08-1560

T. Soussi and K. G. Wiman, Shaping Genetic Alterations in Human Cancer: The p53 Mutation Paradigm, Cancer Cell, vol.12, issue.4, pp.303-312, 2007.
DOI : 10.1016/j.ccr.2007.10.001

B. Vogelstein, D. Lane, and A. J. Levine, Surfing the p53 network, Nature, vol.408, issue.6810, pp.307-310, 2000.
DOI : 10.1038/35042675

A. Petitjean, M. I. Achatz, A. L. Borresen-dale, P. Hainaut, and M. Olivier, TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes, Oncogene, vol.47, issue.15, pp.2157-2165, 2007.
DOI : 10.1158/1078-0432.CCR-05-0507

H. Song, M. Hollstein, and Y. Xu, p53 gain-of-function cancer mutants induce genetic instability by inactivating ATM, Nature Cell Biology, vol.113, issue.5, pp.573-580, 2007.
DOI : 10.1074/jbc.M306938200

D. P. Liu, H. Song, and Y. Xu, A common gain of function of p53 cancer mutants in inducing genetic instability, Oncogene, vol.60, issue.7, pp.949-956, 2010.
DOI : 10.1038/nri1804

T. Soussi, B. Asselain, and D. Hamroun, Meta-analysis of the p53 Mutation Database for Mutant p53 Biological Activity Reveals a Methodologic Bias in Mutation Detection, Clinical Cancer Research, vol.12, issue.1, pp.62-69, 2006.
DOI : 10.1158/1078-0432.CCR-05-0413

URL : https://hal.archives-ouvertes.fr/hal-00018575

T. Soussi, D. Hamroun, and L. Hjortsberg, MUT-TP53 2.0: a novel versatile matrix for statistical analysis of TP53 mutations in human cancera, Human Mutation, vol.358, issue.9, pp.1020-1025, 2010.
DOI : 10.1002/humu.21313

J. Weiss, M. Heine, and K. C. Arden, Mutation and Expression of TP53 in Malignant Melanomas, Recent Results in Cancer Research, vol.139, pp.137-154, 1995.
DOI : 10.1007/978-3-642-78771-3_10

E. Jassem, J. Niklinski, and R. , Types and localisation of p53 gene mutations, Lung Cancer, vol.34, issue.2, pp.47-51, 2001.
DOI : 10.1016/S0169-5002(01)00344-0

X. M. Wu, J. G. Fu, and W. Z. Ge, Screen p53 mutations in hepatocellular carcinoma by FASAY: A novel splicing mutation, Journal of Zhejiang University SCIENCE B, vol.8, issue.2, pp.81-87, 2007.
DOI : 10.1631/jzus.2007.B0081

M. Olivier, A. Langerod, and P. Carrieri, The Clinical Value of Somatic TP53 Gene Mutations in 1,794 Patients with Breast Cancer, Clinical Cancer Research, vol.12, issue.4, pp.1157-1167, 2006.
DOI : 10.1158/1078-0432.CCR-05-1029

L. Lode, M. Eveillard, and V. Trichet, Mutations in TP53 are exclusively associated with del(17p) in multiple myeloma, Haematologica, vol.95, issue.11, pp.1973-1976, 2010.
DOI : 10.3324/haematol.2010.023697

L. Stefancikova, M. Moulis, and P. Fabian, Loss of the p53 tumor suppressor activity is associated with negative prognosis of mantle cell lymphoma, International Journal of Oncology, vol.36, issue.3, pp.699-706, 2010.

L. Pusztai, C. Mazouni, K. Anderson, Y. Wu, and W. F. Symmans, Molecular Classification of Breast Cancer: Limitations and Potential, The Oncologist, vol.11, issue.8, pp.868-877, 2006.
DOI : 10.1634/theoncologist.11-8-868

C. M. Perou, T. Sorile, and M. B. Eisen, Molecular portraits of human breast tumours, Nature, vol.406, issue.6797, pp.747-752, 2000.
DOI : 10.1038/35021093

O. Gluz, C. Liedtke, N. Gottschalk, L. Pusztai, U. Nitz et al., Triple-negative breast cancer--current status and future directions, Annals of Oncology, vol.20, issue.12, pp.1913-1927, 2009.
DOI : 10.1093/annonc/mdp492

URL : http://annonc.oxfordjournals.org/cgi/content/short/20/12/1913

F. C. Bidard, R. Conforti, T. Boulet, S. Michiels, S. Delaloge et al., Does triple-negative phenotype accurately identify basal-like tumour? An immunohistochemical analysis based on 143 'triple-negative' breast cancers, Annals of Oncology, vol.18, issue.7, pp.1285-1286, 2007.
DOI : 10.1093/annonc/mdm360

L. A. Carey, C. M. Perou, and C. A. Livasy, Race, Breast Cancer Subtypes, and Survival in the Carolina Breast Cancer Study, JAMA, vol.295, issue.21, pp.2492-2502, 2006.
DOI : 10.1001/jama.295.21.2492

B. J. Chae, J. S. Bae, and A. Lee, p53 as a Specific Prognostic Factor in Triple-negative Breast Cancer, Japanese Journal of Clinical Oncology, vol.39, issue.4, pp.217-224, 2009.
DOI : 10.1093/jjco/hyp007

M. C. Cheang, D. Voduc, and C. Bajdik, Basal-Like Breast Cancer Defined by Five Biomarkers Has Superior Prognostic Value than Triple-Negative Phenotype, Clinical Cancer Research, vol.14, issue.5, pp.1368-1376, 2008.
DOI : 10.1158/1078-0432.CCR-07-1658

T. Sorlie, C. M. Perou, and R. Tibshirani, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications, Proceedings of the National Academy of Sciences, vol.98, issue.19, pp.10869-10874, 2001.
DOI : 10.1073/pnas.191367098

L. L. Nakopoulou, A. Alexiadou, G. E. Theodoropoulos, A. C. Lazaris, A. Tzonou et al., PROGNOSTIC SIGNIFICANCE OF THE CO-EXPRESSION OF p53 AND c-erbB-2 PROTEINS IN BREAST CANCER, The Journal of Pathology, vol.47, issue.1, pp.31-38, 1996.
DOI : 10.1002/(SICI)1096-9896(199605)179:1<31::AID-PATH523>3.0.CO;2-O

S. E. Singletary, C. Allred, and P. Ashley, Revision of the American Joint Committee on Cancer Staging System for Breast Cancer, Journal of Clinical Oncology, vol.20, issue.17, pp.3628-3636, 2002.
DOI : 10.1200/JCO.2002.02.026

S. Van-laere, I. Van-der-auwera, and G. Van-den-eynden, Distinct molecular phenotype of inflammatory breast cancer compared to non-inflammatory breast cancer using Affymetrix-based genome-wide gene-expression analysis, British Journal of Cancer, vol.5, issue.8, pp.1165-1174, 2007.
DOI : 10.1016/j.biocel.2004.09.006

E. Turpin, I. Bì-eche, and P. Bertheau, Increased incidence of ERBB2 overexpression and TP53 mutation in inflammatory breast cancer, Oncogene, vol.21, issue.49, pp.7593-7597, 2002.
DOI : 10.1038/sj.onc.1205932

M. Sawaki, Y. Ito, and F. Akiyama, High prevalence of HER-2/neu and p53 overexpression in inflammatory breast cancer, Breast Cancer, vol.2, issue.2, pp.172-178, 2006.
DOI : 10.2325/jbcs.13.172

M. Hensel, A. Schneeweiss, and H. P. Sinn, P53 is the strongest predictor of survival in high-risk primary breast cancer patients undergoing high-dose chemotherapy with autologous blood stem cell support, International Journal of Cancer, vol.24, issue.3, pp.290-296, 2002.
DOI : 10.1002/ijc.10478

V. Malamou-mitsi, H. Gogas, and U. Dafni, Evaluation of the prognostic and predictive value of p53 and Bcl-2 in breast cancer patients participating in a randomized study with dose-dense sequential adjuvant chemotherapy, Annals of Oncology, vol.17, issue.10, pp.1504-1511, 2006.
DOI : 10.1093/annonc/mdl147

T. Soussi, p53 alterations in human cancer: more questions than answers, Oncogene, vol.10, issue.15, pp.2145-2156, 2007.
DOI : 10.1006/jmbi.1999.3174

P. Bertheau, F. Plassa, and M. Espié, Effect of mutated TP53 on response of advanced breast cancers to high-dose chemotherapy, The Lancet, vol.360, issue.9336, pp.852-854, 2002.
DOI : 10.1016/S0140-6736(02)09969-5

P. Bertheau, E. Turpin, and D. S. Rickman, Exquisite Sensitivity of TP53 Mutant and Basal Breast Cancers to a Dose-Dense Epirubicin???Cyclophosphamide Regimen, PLoS Medicine, vol.21, issue.3, p.90, 2007.
DOI : 10.1371/journal.pmed.0040090.st009

URL : https://hal.archives-ouvertes.fr/inserm-00700961

J. Lehmann-che, F. André, and C. Desmedt, Cyclophosphamide Dose Intensification May Circumvent Anthracycline Resistance of p53 Mutant Breast Cancers, The Oncologist, vol.15, issue.3, pp.246-252, 2010.
DOI : 10.1634/theoncologist.2009-0243

M. Varna, J. Lehmann-che, and E. Turpin, p53 dependent cell-cycle arrest triggered by chemotherapy in xenografted breast tumors, International Journal of Cancer, vol.104, issue.4, pp.991-997, 2009.
DOI : 10.1002/ijc.24049

URL : https://hal.archives-ouvertes.fr/inserm-00498765

T. Aas, A. L. Borresen, and S. Geisler, Specific P53 mutations are associated with de novo resistance to doxorubicin in breast cancer patients, Nature Medicine, vol.19, issue.7, pp.811-814, 1996.
DOI : 10.1016/0959-8049(94)00213-O

A. L. Borresen, T. I. Andersen, and J. E. Eyfjord, TP53 mutations and breast cancer prognosis: Particularly poor survival rates for cases with mutations in the zinc-binding domains, Genes, Chromosomes and Cancer, vol.53, issue.1, pp.71-75, 1995.
DOI : 10.1002/gcc.2870140113

D. Bergamaschi, M. Gasco, and L. Hiller, p53 polymorphism influences response in cancer chemotherapy via modulation of p73-dependent apoptosis, Cancer Cell, vol.3, issue.4, pp.387-402, 2003.
DOI : 10.1016/S1535-6108(03)00079-5

C. Maisse, P. Guerrieri, and G. Melino, p73 and p63 protein stability: the way to regulate function?, Biochemical Pharmacology, vol.66, issue.8, pp.1555-1561, 2003.
DOI : 10.1016/S0006-2952(03)00511-2

D. Goldschneider, E. Blanc, G. Raguenez, H. Haddada, J. Bénard et al., When p53 needs p73 to be functional ??? forced p73 expression induces nuclear accumulation of endogenous p53 protein, Cancer Letters, vol.197, issue.1-2, pp.99-103, 2003.
DOI : 10.1016/S0304-3835(03)00089-2

Y. Xu, L. Yao, and T. Ouyang, p53 Codon 72 Polymorphism Predicts the Pathologic Response to Neoadjuvant Chemotherapy in Patients with Breast Cancer, Clinical Cancer Research, vol.11, issue.20, pp.7328-7333, 2005.
DOI : 10.1158/1078-0432.CCR-05-0507

F. C. Bidard, M. C. Matthieu, and P. Chollet, p53 status and efficacy of primary anthracyclines/alkylating agent-based regimen according to breast cancer molecular classes, Annals of Oncology, vol.19, issue.7, pp.1261-1265, 2008.
DOI : 10.1093/annonc/mdn039

J. Alsner, V. Jensen, and M. Kyndi, mutations as prognostic variables in tumours from breast cancer patients, Acta Oncologica, vol.88, issue.4, pp.600-607, 2008.
DOI : 10.1056/NEJM199710023371401

W. J. Kostler, T. Brodowicz, and G. Hudelist, The efficacy of trastuzumab in Her-2/neu-overexpressing metastatic breast cancer is independent of p53 status, Journal of Cancer Research and Clinical Oncology, vol.3, issue.7, pp.420-428, 2005.
DOI : 10.1007/s00432-005-0670-3

L. Fernandez-cuesta, S. Anaganti, P. Hainaut, and M. Olivier, p53 status influences response to tamoxifen but not to fulvestrant in breast cancer cell lines, International Journal of Cancer, vol.9, issue.Suppl 1, pp.1813-1821, 2010.
DOI : 10.1002/ijc.25512

E. M. Berns, J. A. Foekens, and R. Vossen, Complete sequencing of TP53 predicts poor response to systemic therapy of advanced breast cancer, Cancer Research, vol.60, issue.8, pp.2155-2162, 2000.

H. S. Kim, C. K. Yom, and H. J. Kim, Overexpression of p53 is correlated with poor outcome in premenopausal women with breast cancer treated with tamoxifen after chemotherapy, Breast Cancer Research and Treatment, vol.126, issue.3, pp.777-788, 2010.
DOI : 10.1007/s10549-009-0560-5

A. Hamilton and M. Piccart, The contribution of molecular markers to the prediction of response in the treatment of breast cancer: a review of the literature on HER-2, p53 and BCL-2, Annals of Oncology, vol.11, issue.6, pp.647-663, 2000.
DOI : 10.1023/A:1008390429428

S. W. Lowe, S. Bodis, and A. Mcclatchey, p53 status and the efficacy of cancer therapy in vivo, Science, vol.266, issue.5186, pp.807-810, 1994.
DOI : 10.1126/science.7973635