C. Mathers, W. Organization, and I. Fleming, World Health Organization Available from: http://www.ipubli.inserm.fr/bitstream/handle, 156 p. 3. 102.pdf [Internet]. Union Internationale Contre le Cancer and the American Joint Committee on Cancer. Cancer, pp.1803-1807, 1997.

G. Locker, S. Hamilton, J. Harris, J. Jessup, N. Kemeny et al., ASCO 2006 Update of Recommendations for the Use of Tumor Markers in Gastrointestinal Cancer, Journal of Clinical Oncology, vol.24, issue.33, pp.5313-5340, 2006.
DOI : 10.1200/JCO.2006.08.2644

A. Prat, J. Parker, O. Karginova, C. Fan, C. Livasy et al., Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer, Breast Cancer Research, vol.15, issue.1, p.68, 2010.
DOI : 10.1038/nm0809-842

M. Ignatiadis and C. Sotiriou, Luminal breast cancer: from biology to treatment, Nature Reviews Clinical Oncology, vol.57, issue.9, 2013.
DOI : 10.1038/nrclinonc.2012.61

C. Livasy, G. Karaca, R. Nanda, M. Tretiakova, O. Olopade et al., Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma, Modern Pathology, vol.193, issue.2, 2006.
DOI : 10.1002/1096-9896(2000)9999:9999<::AID-PATH752>3.0.CO;2-V

E. Jensen and V. Jordan, The estrogen receptor: a model for molecular medicine, Clin Cancer Res Off J Am Assoc Cancer Res, vol.9, issue.6, pp.1980-1989, 2003.

S. Boyd, Remarks on Oöphorectomy in the Treatment of Cancer of the Breast, Br Med J. 1899 Feb, vol.41, pp.257-62, 1988.

E. Allen, E. Doisy, E. A. Allen, . Doisy, and . Jama, Landmark article Sept 8 An ovarian hormone. Preliminary report on its localization, extraction and partial purification, and action in test animals, pp.2681-2684, 1923.

E. Jensen, G. Block, S. Smith, K. Kyser, and E. Desombre, Estrogen receptors and breast cancer response to adrenalectomy. Natl Cancer Inst Monogr, pp.55-70, 1971.

K. Horwitz and W. Mcguire, Specific progesterone receptors in human breast cancer, Steroids, vol.25, issue.4, 1975.
DOI : 10.1016/0039-128X(75)90027-6

S. Ogawa, S. Inoue, T. Watanabe, H. Hiroi, A. Orimo et al., The complete primary structure of human estrogen receptor beta (hER beta) and its heterodimerization with ER alpha in vivo and in vitro, Biochem Biophys Res Commun Feb, vol.4243, issue.1, pp.122-128, 1998.

G. Kerdivel, G. Flouriot, and F. Pakdel, Modulation of Estrogen Receptor Alpha Activity and Expression During Breast Cancer Progression, Vitam Horm, vol.93, pp.135-60, 2013.
DOI : 10.1016/B978-0-12-416673-8.00004-6

URL : https://hal.archives-ouvertes.fr/hal-00874313

H. Goulding, S. Pinder, P. Cannon, D. Pearson, R. Nicholson et al., A new immunohistochemical antibody for the assessment of estrogen receptor status on routine formalin-fixed tissue samples, Human Pathology, vol.26, issue.3, pp.291-295, 1995.
DOI : 10.1016/0046-8177(95)90060-8

J. Shaw, K. Udokang, J. Mosquera, H. Chauhan, J. Jones et al., Oestrogen receptors alpha and beta differ in normal human breast and breast carcinomas, The Journal of Pathology, vol.97, issue.4, pp.450-457, 2002.
DOI : 10.1038/bjc.1992.122

L. Pertschuk, J. Feldman, Y. Kim, L. Braithwaite, F. Schneider et al., Estrogen receptor immunocytochemistry in paraffin embedded tissues with ER1D5 predicts breast cancer endocrine response more accurately than H222Sp gamma in frozen sections or cytosol-based ligand-binding assays. Cancer, pp.2514-2523, 1996.

D. Barnes, W. Harris, P. Smith, R. Millis, and R. Rubens, Immunohistochemical determination of oestrogen receptor: comparison of different methods of assessment of staining and correlation with clinical outcome of breast cancer patients, British Journal of Cancer, vol.74, issue.9, pp.1445-51, 1996.
DOI : 10.1038/bjc.1996.563

L. Diaz and N. Sneige, Estrogen Receptor Analysis for Breast Cancer, Advances in Anatomic Pathology, vol.12, issue.1, pp.10-19, 2005.
DOI : 10.1097/00125480-200501000-00003

M. Cheang, D. Treaba, C. Speers, I. Olivotto, C. Bajdik et al., Immunohistochemical Detection Using the New Rabbit Monoclonal Antibody SP1 of Estrogen Receptor in Breast Cancer Is Superior to Mouse Monoclonal Antibody 1D5 in Predicting Survival, Journal of Clinical Oncology, vol.24, issue.36, pp.5637-5681, 2006.
DOI : 10.1200/JCO.2005.05.4155

P. Kastner, A. Krust, B. Turcotte, U. Stropp, L. Tora et al., Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B, EMBO J, vol.9, issue.5, pp.1603-1617, 1990.

J. Condon, D. Hardy, K. Kovaric, and C. Mendelson, Up-Regulation of the Progesterone Receptor (PR)-C Isoform in Laboring Myometrium by Activation of Nuclear Factor-??B May Contribute to the Onset of Labor through Inhibition of PR Function, Molecular Endocrinology, vol.20, issue.4, pp.764-75, 2006.
DOI : 10.1210/me.2005-0242

M. Salazar, A. Lerma-ortiz, G. Hooks, A. Ashley, and R. Ashley, Progestin-mediated activation of MAPK and AKT in nuclear progesterone receptor negative breast epithelial cells: The role of membrane progesterone receptors, Gene, vol.591, issue.1, pp.6-13, 2016.
DOI : 10.1016/j.gene.2016.06.044

F. Blows, K. Driver, M. Schmidt, A. Broeks, F. 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 Medicine, vol.12, issue.5, p.1000279, 2010.
DOI : 10.1371/journal.pmed.1000279.s011

F. Vignon, S. Bardon, D. Chalbos, and H. Rochefort, Antiestrogenic Effect of R5020, a Synthetic Progestin in Human Breast Cancer Cells in Culture*, The Journal of Clinical Endocrinology & Metabolism, vol.56, issue.6, pp.1124-1154, 1983.
DOI : 10.1210/jcem-56-6-1124

E. Musgrove, A. Swarbrick, C. Lee, A. Cornish, and R. Sutherland, Mechanisms of Cyclin-Dependent Kinase Inactivation by Progestins, Molecular and Cellular Biology, vol.18, issue.4, pp.1812-1837, 1998.
DOI : 10.1128/MCB.18.4.1812

C. Chen, D. Hardy, and C. Mendelson, Progesterone Receptor Inhibits Proliferation of Human Breast Cancer Cells via Induction of MAPK Phosphatase 1 (MKP-1/DUSP1), Journal of Biological Chemistry, vol.169, issue.50, pp.43091-102, 2011.
DOI : 10.1074/jbc.M109.021469

P. Kabos, J. Finlay-schultz, C. Li, E. Kline, C. Finlayson et al., Patient-derived luminal breast cancer xenografts retain hormone receptor heterogeneity and help define unique estrogen-dependent gene signatures, Breast Cancer Research and Treatment, vol.7, issue.4, pp.415-447, 2012.
DOI : 10.1371/journal.pone.0035859

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3818141/pdf

H. Mohammed, I. Russell, R. Stark, O. Rueda, T. Hickey et al., Progesterone receptor modulates ER?? action in breast cancer, Nature, vol.27, issue.7560, pp.313-320, 2015.
DOI : 10.1200/JCO.2008.18.1370

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4650274/pdf

V. Bardou, G. Arpino, R. Elledge, C. Osborne, and G. Clark, Progesterone Receptor Status Significantly Improves Outcome Prediction Over Estrogen Receptor Status Alone for Adjuvant Endocrine Therapy in Two Large Breast Cancer Databases, Journal of Clinical Oncology, vol.21, issue.10, pp.1973-1982, 2003.
DOI : 10.1200/JCO.2003.09.099

M. Press, B. Spaulding, S. Groshen, D. Kaminsky, M. Hagerty et al., Comparison of different antibodies for detection of progesterone receptor in breast cancer, Steroids, vol.67, issue.9, pp.799-813, 2002.
DOI : 10.1016/S0039-128X(02)00039-9

T. Holbro, G. Civenni, and N. Hynes, The ErbB receptors and their role in cancer progression, Experimental Cell Research, vol.284, issue.1, pp.99-110, 2003.
DOI : 10.1016/S0014-4827(02)00099-X

T. Holbro and N. Hynes, : Directing Key Signaling Networks Throughout Life, Annual Review of Pharmacology and Toxicology, vol.44, issue.1, pp.195-217, 2004.
DOI : 10.1146/annurev.pharmtox.44.101802.121440

A. Gschwind, O. Fischer, and A. Ullrich, Timeline: The discovery of receptor tyrosine kinases: targets for cancer therapy, Nature Reviews Cancer, vol.4, issue.5, pp.361-70, 2004.
DOI : 10.1038/nrc1360

J. Couturier, A. Vincent-salomon, A. Nicolas, P. Beuzeboc, E. Mouret et al., Strong Correlation between Results of Fluorescent In Situ Hybridization and Immunohistochemistry for the Assessment of the ERBB2 (HER-2/neu) Gene Status in Breast Carcinoma, Modern Pathology, vol.10, issue.Suppl. 1, 2000.
DOI : 10.1002/gcc.2870100303

T. Sorlie, R. Tibshirani, J. Parker, T. Hastie, J. Marron et al., Repeated observation of breast tumor subtypes in independent gene expression data sets, Proceedings of the National Academy of Sciences, vol.360, issue.9338, pp.8418-8441, 2003.
DOI : 10.1016/S0140-6736(02)11087-7

T. Sørlie, C. Perou, R. Tibshirani, T. Aas, S. Geisler et al., Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications, Proceedings of the National Academy of Sciences, vol.179, issue.1, pp.10869-74, 2001.
DOI : 10.1002/(SICI)1096-9896(199605)179:1<31::AID-PATH523>3.0.CO;2-O

D. Hicks and S. Kulkarni, HER2+ Breast Cancer, American Journal of Clinical Pathology, vol.129, issue.2, pp.263-73, 2008.
DOI : 10.1309/99AE032R9FM8WND1

J. Gerdes, U. Schwab, H. Lemke, and H. Stein, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation, International Journal of Cancer, vol.18, issue.1, pp.13-20, 1983.
DOI : 10.1177/28.8.7003001

J. Gerdes, H. Lemke, H. Baisch, H. Wacker, U. Schwab et al., Cell cycle analysis of a cell proliferationassociated human nuclear antigen defined by the monoclonal antibody Ki-67, J Immunol Baltim Md, vol.133, issue.4, pp.1710-1715, 1950.

C. Schlüter, M. Duchrow, C. Wohlenberg, M. Becker, G. Key et al., The cell proliferation-associated antigen of antibody Ki-67: a very large, ubiquitous nuclear protein with numerous repeated elements, representing a new kind of cell cycle-maintaining proteins, The Journal of Cell Biology, vol.123, issue.3, pp.513-535, 1993.
DOI : 10.1083/jcb.123.3.513

T. Scholzen and J. Gerdes, The Ki-67 protein: From the known and the unknown, Journal of Cellular Physiology, vol.19, issue.31, pp.311-333, 2000.
DOI : 10.1093/ajcp/110.1.24

A. Urruticoechea, I. Smith, and M. Dowsett, Proliferation Marker Ki-67 in Early Breast Cancer, Journal of Clinical Oncology, vol.23, issue.28, pp.7212-7232, 2005.
DOI : 10.1200/JCO.2005.07.501

E. De-azambuja, F. Cardoso, G. De-castro, M. Colozza, M. Mano et al., Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12???155 patients, British Journal of Cancer, vol.27, issue.10, pp.1504-1517, 2007.
DOI : 10.1002/1097-0142(19910115)67:2<421::AID-CNCR2820670217>3.0.CO;2-Q

R. Yerushalmi, R. Woods, P. Ravdin, M. Hayes, and K. Gelmon, Ki67 in breast cancer: prognostic and predictive potential, The Lancet Oncology, vol.11, issue.2, pp.174-83, 2010.
DOI : 10.1016/S1470-2045(09)70262-1

R. Stuart-harris, C. Caldas, S. Pinder, and P. Pharoah, Proliferation markers and survival in early breast cancer: A systematic review and meta-analysis of 85 studies in 32,825 patients, The Breast, vol.17, issue.4, 2008.
DOI : 10.1016/j.breast.2008.02.002

A. Goldhirsch, W. Wood, A. Coates, R. Gelber, B. Thürlimann et al., Strategies for subtypes-dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer, Ann Oncol Off J Eur Soc Med Oncol, 2011.

A. Goldhirsch, E. Winer, A. Coates, R. Gelber, M. Piccart-gebhart et al., Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013, Annals of Oncology, vol.24, issue.9, pp.2206-2229, 2013.
DOI : 10.1093/annonc/mdt303

A. Goldhirsch, J. Ingle, R. Gelber, A. Coates, B. Thürlimann et al., Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2009, Annals of Oncology, vol.20, issue.8, pp.1319-1348, 2009.
DOI : 10.1093/annonc/mdp322

C. Denkert, J. Budczies, G. Von-minckwitz, S. Wienert, S. Loibl et al., Strategies for developing Ki67 as a useful biomarker in breast cancer, The Breast, vol.24, issue.2, pp.67-72, 2015.
DOI : 10.1016/j.breast.2015.07.017

M. Duffy, The Urokinase Plasminogen Activator System: Role in Malignancy, Current Pharmaceutical Design, vol.10, issue.1, pp.39-49, 2004.
DOI : 10.2174/1381612043453559

K. Danø, N. Behrendt, G. Høyer-hansen, M. Johnsen, L. Lund et al., Plasminogen activation and cancer, Thrombosis and Haemostasis, vol.93, issue.4, pp.676-81, 2005.
DOI : 10.1160/TH05-01-0054

M. Schmitt, K. Mengele, R. Napieralski, V. Magdolen, U. Reuning et al., Clinical utility of level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1, Expert Review of Molecular Diagnostics, vol.10, issue.8, pp.1051-67, 2010.
DOI : 10.1586/erm.10.73

E. Deryugina and J. Quigley, Cell Surface Remodeling by Plasmin: A New Function for an Old Enzyme, Journal of Biomedicine and Biotechnology, vol.41, issue.7, p.564259, 2012.
DOI : 10.1042/BJ20110129

URL : http://doi.org/10.1155/2012/564259

M. Tjwa, R. Moura, L. Moons, S. Plaisance, D. Mol et al., Fibrinolysis-independent role of plasmin and its activators in the haematopoietic recovery after myeloablation, Journal of Cellular and Molecular Medicine, vol.22, issue.11-12, 2009.
DOI : 10.1161/01.RES.81.5.829

G. Ferraris and N. Sidenius, Urokinase plasminogen activator receptor: a functional integrator of extracellular proteolysis, cell adhesion, and signal transduction, Semin Thromb Hemost, 2013.

M. Jo, K. Thomas, N. Marozkina, T. Amin, C. Silva et al., Dynamic Assembly of the Urokinase-type Plasminogen Activator Signaling Receptor Complex Determines the Mitogenic Activity of Urokinase-type Plasminogen Activator, Journal of Biological Chemistry, vol.16, issue.17, pp.17449-57, 2005.
DOI : 10.1074/jbc.M101303200

J. Kiyan, R. Kiyan, H. Haller, and I. Dumler, Urokinase-induced signaling in human vascular smooth muscle cells is mediated by PDGFR-??, The EMBO Journal, vol.271, issue.10, pp.1787-97, 2005.
DOI : 10.1128/MCB.19.5.3727

URL : http://embojnl.embopress.org/content/embojnl/24/10/1787.full.pdf

N. Montuori, V. Cosimato, L. Rinaldi, V. Rea, D. Alfano et al., uPAR regulates pericellular proteolysis through a mechanism involving integrins and fMLF-receptors, Thrombosis and Haemostasis, vol.109, issue.2, 2013.
DOI : 10.1160/TH12-08-0546

D. Dupont, J. Madsen, T. Kristensen, J. Bodker, G. Blouse et al., Biochemical properties of plasminogen activator inhibitor-1. Front Biosci Landmark Ed, pp.1337-61, 2009.
DOI : 10.2741/3312

A. Nykjaer, M. Conese, E. Christensen, D. Olson, O. Cremona et al., Recycling of the urokinase receptor upon internalization of the uPA:serpin complexes, The EMBO Journal, vol.16, issue.10, pp.2610-2630, 1997.
DOI : 10.1093/emboj/16.10.2610

T. Bugge, L. Lund, K. Kombrinck, B. Nielsen, K. Holmbäck et al., Reduced metastasis of Polyoma virus middle T antigen-induced mammary cancer in plasminogen-deficient mice. Oncogene, pp.3097-104, 1998.

E. Bekes, E. Deryugina, T. Kupriyanova, E. Zajac, K. Botkjaer et al., Activation of Pro-uPA Is Critical for Initial Escape from the Primary Tumor and Hematogenous Dissemination of Human Carcinoma Cells, Neoplasia, vol.13, issue.9, pp.806-827, 2011.
DOI : 10.1593/neo.11704

T. Masuda, N. Hattori, T. Senoo, S. Akita, N. Ishikawa et al., SK-216, an Inhibitor of Plasminogen Activator Inhibitor-1, Limits Tumor Progression and Angiogenesis, Molecular Cancer Therapeutics, vol.12, issue.11, 2013.
DOI : 10.1158/1535-7163.MCT-13-0041

URL : http://mct.aacrjournals.org/content/molcanther/12/11/2378.full.pdf

H. Fang, V. Placencio, and Y. Declerck, Protumorigenic Activity of Plasminogen Activator Inhibitor-1 Through an Antiapoptotic Function, JNCI: Journal of the National Cancer Institute, vol.104, issue.19, pp.1470-84, 2012.
DOI : 10.1093/jnci/djs377

M. Duffy, O. Grady, P. Devaney, D. O-'siorain, L. Fennelly et al., Urokinase-plasminogen activator, a marker for aggressive breast carcinomas. Preliminary report. Cancer, pp.531-534, 1988.
DOI : 10.1002/1097-0142(19880801)62:3<531::aid-cncr2820620315>3.0.co;2-b

URL : http://onlinelibrary.wiley.com/doi/10.1002/1097-0142(19880801)62:3<531::AID-CNCR2820620315>3.0.CO;2-B/pdf

N. Harbeck, R. Kates, and M. Schmitt, Clinical relevance of invasion factors urokinase-type plasminogen activator and plasminogen activator inhibitor type 1 for individualized therapy decisions in primary breast cancer is greatest when used in combination, J Clin Oncol Off J Am Soc Clin Oncol Feb, vol.1520, issue.4, pp.1000-1007, 2002.

N. Harbeck, R. Kates, M. Look, M. Meijer-van-gelder, J. Klijn et al., Enhanced benefit from adjuvant chemotherapy in breast cancer patients classified high-risk according to urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor type 1 (n = 3424). Cancer Res, pp.4617-4639, 2002.

P. Manders, V. Tjan-heijnen, P. Span, N. Grebenchtchikov, J. Foekens et al., Predictive Impact of Urokinase-Type Plasminogen Activator, Cancer Research, vol.64, issue.2, pp.659-64, 2004.
DOI : 10.1158/0008-5472.CAN-03-1820

URL : http://cancerres.aacrjournals.org/content/canres/64/2/659.full.pdf

S. Borstnar, A. Sadikov, B. Mozina, and T. Cufer, High levels of uPA and PAI-1 predict a good response to anthracyclines, Breast Cancer Research and Treatment, vol.21, issue.6, pp.615-639, 2010.
DOI : 10.1200/JCO.2006.01.903

URL : https://hal.archives-ouvertes.fr/hal-00612980

L. Veer, H. Dai, M. Van-de-vijver, Y. He, A. Hart et al., Gene expression profiling predicts clinical outcome of breast cancer, Nature, vol.344, issue.6871, pp.530-536, 2002.
DOI : 10.1056/NEJM200102223440801

K. Mir and E. Southern, Erratum: Determining the influence of structure on hybridization using oligonucleotide arrays, Nature Biotechnology, vol.17, issue.10, pp.788-92, 1999.
DOI : 10.1038/13726

M. Filipits, M. Rudas, R. Jakesz, P. Dubsky, F. Fitzal et al., A New Molecular Predictor of Distant Recurrence in ER-Positive, HER2-Negative Breast Cancer Adds Independent Information to Conventional Clinical Risk Factors, Clinical Cancer Research, vol.17, issue.18, pp.6012-6032, 2011.
DOI : 10.1158/1078-0432.CCR-11-0926

S. Paik, S. Shak, G. Tang, C. Kim, J. Baker et al., A Multigene Assay to Predict Recurrence of Tamoxifen-Treated, Node-Negative Breast Cancer, New England Journal of Medicine, vol.351, issue.27, pp.2817-2843, 2004.
DOI : 10.1056/NEJMoa041588

B. Fisher, J. Costantino, C. Redmond, R. Poisson, D. Bowman et al., A randomized clinical trial evaluating tamoxifen in the treatment of patients with node-negative breast cancer who have estrogenreceptor-positive tumors, N Engl J Med Feb, vol.23320, issue.8, pp.479-84, 1989.

B. Fisher, J. Jeong, J. Bryant, S. Anderson, J. Dignam et al., Treatment of lymph-node-negative, oestrogen-receptor-positive breast cancer: long-term findings from National Surgical Adjuvant Breast and Bowel Project randomised clinical trials, The Lancet, vol.364, issue.9437, pp.858-68, 2004.
DOI : 10.1016/S0140-6736(04)16981-X

B. Fisher, J. Dignam, N. Wolmark, A. Decillis, B. Emir et al., Tamoxifen and Chemotherapy for Lymph Node-Negative, Estrogen Receptor-Positive Breast Cancer, JNCI Journal of the National Cancer Institute, vol.89, issue.22, pp.1673-82, 1997.
DOI : 10.1093/jnci/89.22.1673

URL : https://academic.oup.com/jnci/article-pdf/89/22/1673/7708589/89-22-1673.pdf

S. Paik, G. Tang, S. Shak, C. Kim, J. Baker et al., Gene Expression and Benefit of Chemotherapy in Women With Node-Negative, Estrogen Receptor???Positive Breast Cancer, Journal of Clinical Oncology, vol.24, issue.23, pp.3726-3760, 2006.
DOI : 10.1200/JCO.2005.04.7985

B. Gy?rffy, C. Hatzis, T. Sanft, E. Hofstatter, B. Aktas et al., Multigene prognostic tests in breast cancer: past, present, future, Breast Cancer Research, vol.26, issue.24 Suppl, p.11, 2015.
DOI : 10.1200/JCO.2007.14.4147

S. Mook, M. Schmidt, G. Viale, G. Pruneri, I. Eekhout et al., The 70-gene prognosis-signature predicts disease outcome in breast cancer patients with 1???3 positive lymph nodes in an independent validation study, Breast Cancer Research and Treatment, vol.3, issue.2, pp.295-302, 2009.
DOI : 10.1093/jnci/94.2.116

URL : https://hal.archives-ouvertes.fr/hal-00478252

J. Parker, M. Mullins, M. Cheang, S. Leung, D. Voduc et al., Supervised Risk Predictor of Breast Cancer Based on Intrinsic Subtypes, Journal of Clinical Oncology, vol.27, issue.8, pp.1160-1167, 2009.
DOI : 10.1200/JCO.2008.18.1370

M. Dowsett, I. Sestak, E. Lopez-knowles, K. Sidhu, A. Dunbier et al., DX and IHC4 for Predicting Risk of Distant Recurrence After Endocrine Therapy, Journal of Clinical Oncology, vol.31, issue.22, pp.312783-90, 2013.
DOI : 10.1200/JCO.2012.46.1558

C. Sotiriou, P. Wirapati, S. Loi, A. Harris, S. Fox et al., Gene Expression Profiling in Breast Cancer: Understanding the Molecular Basis of Histologic Grade To Improve Prognosis, JNCI: Journal of the National Cancer Institute, vol.98, issue.4, pp.262-72, 2006.
DOI : 10.1093/jnci/djj052

M. Van-de-vijver, Y. He, . Van-'t, L. Veer, H. Dai et al., A Gene-Expression Signature as a Predictor of Survival in Breast Cancer, New England Journal of Medicine, vol.347, issue.25, pp.1999-2009, 2002.
DOI : 10.1056/NEJMoa021967

A. Prat, J. Parker, C. Fan, M. Cheang, L. Miller et al., Concordance among gene expression-based predictors for ER-positive breast cancer treated with adjuvant tamoxifen, Annals of Oncology, vol.23, issue.11, pp.2866-73, 2012.
DOI : 10.1093/annonc/mds080

URL : https://academic.oup.com/annonc/article-pdf/23/11/2866/13786106/mds080.pdf

C. Kelly, P. Bernard, S. Krishnamurthy, B. Wang, M. Ebbert et al., Agreement in Risk Prediction Between the 21-Gene Recurrence Score Assay (Oncotype DX(R)) and the PAM50 Breast Cancer Intrinsic Classifier?? in Early-Stage Estrogen Receptor-Positive Breast Cancer, The Oncologist, vol.17, issue.4, pp.492-500, 2012.
DOI : 10.1634/theoncologist.2012-0007

T. Iwamoto, J. Lee, G. Bianchini, R. Hubbard, E. Young et al., First generation prognostic gene signatures for breast cancer predict both survival and chemotherapy sensitivity and identify overlapping patient populations, Breast Cancer Research and Treatment, vol.9, issue.6, pp.155-64, 2011.
DOI : 10.1186/1471-2164-9-394

J. Cuzick, M. Dowsett, S. Pineda, C. Wale, J. Salter et al., Prognostic Value of a Combined Estrogen Receptor, Progesterone Receptor, Ki-67, and Human Epidermal Growth Factor Receptor 2 Immunohistochemical Score and Comparison With the Genomic Health Recurrence Score in Early Breast Cancer, Journal of Clinical Oncology, vol.29, issue.32, pp.4273-4281, 2011.
DOI : 10.1200/JCO.2010.31.2835

C. Bonneau, Z. Gurard-levin, F. Andre, L. Pusztai, and R. Rouzier, Predictive and Prognostic Value of the TauProtein in Breast Cancer, Anticancer Res, vol.35, issue.10, pp.5179-84, 2015.

H. Kim, J. Koh, Y. Choi, J. Ro, H. Kim et al., Chromatin CKAP2, a New Proliferation Marker, as Independent Prognostic Indicator in Breast Cancer, PLoS ONE, vol.134, issue.6, p.98160, 2014.
DOI : 10.1371/journal.pone.0098160.t004

E. Collie-duguid, K. Sweeney, K. Stewart, I. Miller, E. Smyth et al., SerpinB3, a new prognostic tool in breast cancer patients treated with neoadjuvant chemotherapy, Breast Cancer Research and Treatment, vol.7, issue.3, 2012.
DOI : 10.1038/nrclinonc.2010.61

Y. Lin, F. Liu, Y. Fan, X. Qian, R. Lang et al., Both high expression of pyruvate kinase M2 and vascular endothelial growth factor-C predicts poorer prognosis in human breast cancer, Int J Clin Exp Pathol, vol.8, issue.7, pp.8028-8065, 2015.

A. Bozorgi, M. Khazaei, and M. Khazaei, New Findings on Breast Cancer Stem Cells: A Review, Journal of Breast Cancer, vol.18, issue.4, pp.303-315, 2015.
DOI : 10.4048/jbc.2015.18.4.303

URL : http://synapse.koreamed.org/Synapse/Data/PDFData/0096JBC/jbc-18-303.pdf

C. Ginestier, M. Hur, E. Charafe-jauffret, F. Monville, J. Dutcher et al., ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome, Cell Stem Cell, vol.1, issue.5, pp.555-67, 2007.
DOI : 10.1016/j.stem.2007.08.014

URL : https://hal.archives-ouvertes.fr/hal-01431968

B. Lewis, C. Burge, and D. Bartel, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell, pp.15-20, 2005.

R. Friedman, K. Farh, C. Burge, and D. Bartel, Most mammalian mRNAs are conserved targets of microRNAs, Genome Research, vol.19, issue.1, pp.92-105, 2009.
DOI : 10.1101/gr.082701.108

URL : http://genome.cshlp.org/content/19/1/92.full.pdf

A. Kasinski and F. Slack, MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy, Nature Reviews Cancer, vol.39, issue.12, pp.849-64, 2011.
DOI : 10.1038/ng2003

C. Croce, Causes and consequences of microRNA dysregulation in cancer, Nature Reviews Genetics, vol.124, issue.10, pp.704-718, 2009.
DOI : 10.4161/cc.7.6.5834

R. Garzon, G. Calin, and C. Croce, MicroRNAs in Cancer, Annual Review of Medicine, vol.60, issue.1, pp.167-79, 2009.
DOI : 10.1146/annurev.med.59.053006.104707

M. Cortez, C. Ivan, P. Zhou, X. Wu, M. Ivan et al., microRNAs in Cancer, Adv Cancer Res, vol.108, pp.113-57, 2010.
DOI : 10.1016/B978-0-12-380888-2.00004-2

S. Mukherji, M. Ebert, G. Zheng, J. Tsang, P. Sharp et al., MicroRNAs can generate thresholds in target gene expression, Nature Genetics, vol.43, issue.9, pp.854-863, 2011.
DOI : 10.1126/science.1137999

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3163764/pdf

D. Luo, J. Wilson, N. Harvel, J. Liu, L. Pei et al., A systematic evaluation of miRNA:mRNA interactions involved in the migration and invasion of breast cancer cells, Journal of Translational Medicine, vol.11, issue.1, p.57, 2013.
DOI : 10.1016/j.tcb.2010.01.001

C. Welch, Y. Chen, and R. Stallings, MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene, pp.5017-5039, 2007.
DOI : 10.1038/sj.onc.1210293

URL : http://www.nature.com/onc/journal/v26/n34/pdf/1210293a.pdf

F. Chen and S. Hu, Effect of microRNA-34a in cell cycle, differentiation, and apoptosis: A review, Journal of Biochemical and Molecular Toxicology, vol.115, issue.2, pp.79-86, 2012.
DOI : 10.1016/S0092-8674(03)01018-3

M. Kaller, S. Liffers, S. Oeljeklaus, K. Kuhlmann, S. Röh et al., Genome-wide characterization of miR-34a induced changes in protein and mRNA expression by a combined pulsed SILAC and microarray analysis, Mol Cell Proteomics MCP, vol.10, issue.8, pp.111-010462, 2011.

M. Mackiewicz, K. Huppi, J. Pitt, T. Dorsey, S. Ambs et al., Identification of the receptor tyrosine kinase AXL in breast cancer as a target for the human miR-34a microRNA, Breast Cancer Research and Treatment, vol.17, issue.11, pp.663-79, 2011.
DOI : 10.1038/cdd.2009.56

D. Pramanik, N. Campbell, C. Karikari, R. Chivukula, O. Kent et al., Restitution of Tumor Suppressor MicroRNAs Using a Systemic Nanovector Inhibits Pancreatic Cancer Growth in Mice, Molecular Cancer Therapeutics, vol.10, issue.8, pp.1470-80, 2011.
DOI : 10.1158/1535-7163.MCT-11-0152

A. Kasinski and F. Slack, miRNA-34 Prevents Cancer Initiation and Progression in a Therapeutically Resistant K-ras and p53-Induced Mouse Model of Lung Adenocarcinoma, Cancer Research, vol.72, issue.21, pp.5576-87, 2012.
DOI : 10.1158/0008-5472.CAN-12-2001

B. Kumar, A. Yadav, J. Lang, T. Teknos, and P. Kumar, Dysregulation of MicroRNA-34a Expression in Head and Neck Squamous Cell Carcinoma Promotes Tumor Growth and Tumor Angiogenesis, PLoS ONE, vol.85, issue.5, p.37601, 2012.
DOI : 10.1371/journal.pone.0037601.s003

S. Yang, Y. Li, J. Gao, T. Zhang, S. Li et al., MicroRNA-34 suppresses breast cancer invasion and metastasis by directly targeting Fra-1. Oncogene, pp.4294-303, 2013.
DOI : 10.1038/onc.2012.432

L. Li, L. Yuan, J. Luo, J. Gao, J. Guo et al., MiR-34a inhibits proliferation and migration of breast cancer through down-regulation of Bcl-2 and SIRT1, Clinical and Experimental Medicine, vol.17, issue.9, pp.109-126, 2013.
DOI : 10.1158/1078-0432.CCR-10-3244

A. Javeri, M. Ghaffarpour, M. Taha, and M. Houshmand, Downregulation of miR-34a in breast tumors is not associated with either p53 mutations or promoter hypermethylation while it correlates with metastasis, Medical Oncology, vol.377, issue.1, p.413, 2013.
DOI : 10.1016/j.bbrc.2008.09.086

S. Agarwal, J. Hanna, M. Sherman, J. Figueroa, and D. Rimm, Quantitative assessment of miR34a as an independent prognostic marker in breast cancer, British Journal of Cancer, vol.2, issue.1, pp.61-69, 2015.
DOI : 10.1038/onc.2012.432

H. Peurala, D. Greco, T. Heikkinen, S. Kaur, J. Bartkova et al., MiR-34a Expression Has an Effect for Lower Risk of Metastasis and Associates with Expression Patterns Predicting Clinical Outcome in Breast Cancer, PLoS ONE, vol.69, issue.2, p.26122, 2011.
DOI : 10.1371/journal.pone.0026122.s004

E. Van-schooneveld, H. Wildiers, I. Vergote, P. Vermeulen, L. Dirix et al., Dysregulation of microRNAs in breast cancer and their potential role as prognostic and predictive biomarkers in patient management Available from, Breast Cancer Res BCR [Internet], vol.17, 2015.

T. Boon, J. Cerottini, B. Van-den-eynde, P. Van-der-bruggen, and A. Van-pel, Tumor Antigens Recognized by T Lymphocytes, Annual Review of Immunology, vol.12, issue.1, pp.337-65, 1994.
DOI : 10.1146/annurev.iy.12.040194.002005

P. Van-der-bruggen, C. Traversari, P. Chomez, C. Lurquin, D. Plaen et al., A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma, Science, vol.254, issue.5038, pp.1643-1650, 1991.
DOI : 10.1126/science.1840703

V. Shankaran, H. Ikeda, A. Bruce, J. White, P. Swanson et al., IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity, Nature, vol.410, issue.6832, pp.1107-1118, 2001.
DOI : 10.1038/35074122

J. Galon, A. Costes, F. Sanchez-cabo, A. Kirilovsky, B. Mlecnik et al., Type, Density, and Location of Immune Cells Within Human Colorectal Tumors Predict Clinical Outcome, Science, vol.313, issue.5795, pp.1960-1964, 2006.
DOI : 10.1126/science.1129139

P. Baier, S. Wimmenauer, T. Hirsch, V. Specht, B. Von-kleist et al., Analysis of the T Cell Receptor Variability of Tumor-Infiltrating Lymphocytes in Colorectal Carcinomas, Tumor Biology, vol.19, issue.3, pp.205-217, 1998.
DOI : 10.1159/000030008

A. Diederichsen, J. Hjelmborg, B. Christensen, P. Zeuthen, J. Fenger et al., Prognostic value of the CD4+/CD8+ ratio of tumour infiltrating lymphocytes in colorectal cancer and HLA-DR expression on tumour cells, Cancer Immunology, Immunotherapy, vol.52, issue.7, pp.423-431, 2003.
DOI : 10.1007/s00262-003-0388-5

Y. Naito, K. Saito, K. Shiiba, A. Ohuchi, K. Saigenji et al., CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res, pp.3491-3495, 1998.

B. Mlecnik, M. Tosolini, A. Kirilovsky, A. Berger, G. Bindea et al., Histopathologic-Based Prognostic Factors of Colorectal Cancers Are Associated With the State of the Local Immune Reaction, Journal of Clinical Oncology, vol.29, issue.6, pp.610-618, 2011.
DOI : 10.1200/JCO.2010.30.5425

H. Angell and J. Galon, From the immune contexture to the Immunoscore: the role of prognostic and predictive immune markers in cancer, Current Opinion in Immunology, vol.25, issue.2, pp.261-268
DOI : 10.1016/j.coi.2013.03.004

F. Pagès, A. Kirilovsky, B. Mlecnik, M. Asslaber, M. Tosolini et al., In Situ Cytotoxic and Memory T Cells Predict Outcome in Patients With Early-Stage Colorectal Cancer, Journal of Clinical Oncology, vol.27, issue.35, pp.5944-51, 2009.
DOI : 10.1200/JCO.2008.19.6147

J. Galon, B. Mlecnik, G. Bindea, H. Angell, A. Berger et al., Towards the introduction of the ???Immunoscore??? in the classification of malignant tumours, The Journal of Pathology, vol.39, issue.2, pp.199-209
DOI : 10.1016/j.immuni.2013.10.003

T. Puvirajesinghe, F. Bertucci, A. Jain, P. Scerbo, E. Belotti et al., Identification of p62/SQSTM1 as a component of non-canonical Wnt VANGL2???JNK signalling in breast cancer, Nature Communications, vol.14, p.10318, 2016.
DOI : 10.1016/j.cub.2004.05.051

URL : https://hal.archives-ouvertes.fr/hal-01447570

A. Daulat, J. Borg, and . Wnt, Planar Cell Polarity Signaling: New Opportunities for Cancer Treatment. Trends Cancer, Feb, vol.13, issue.2, pp.113-138, 2017.
DOI : 10.1016/j.trecan.2017.01.001

J. Anastas and R. Moon, WNT signalling pathways as therapeutic targets in cancer, Nature Reviews Cancer, vol.284, issue.1, pp.11-26
DOI : 10.1074/jbc.M109.048884

J. Anastas, T. Biechele, M. Robitaille, J. Muster, K. Allison et al., A protein complex of SCRIB, NOS1AP and VANGL1 regulates cell polarity and migration, and is associated with breast cancer progression, Oncogene, vol.365, issue.32, pp.3696-708, 2009.
DOI : 10.1016/j.cell.2008.09.045

T. Gujral, M. Chan, L. Peshkin, P. Sorger, M. Kirschner et al., A noncanonical Frizzled2 pathway regulates epithelial-mesenchymal transition and metastasis. Cell, pp.844-56, 2014.
DOI : 10.1016/j.cell.2014.10.032

URL : https://doi.org/10.1016/j.cell.2014.10.032

J. Hatakeyama, J. Wald, I. Printsev, H. Ho, and K. Carraway, Vangl1 and Vangl2: planar cell polarity components with a developing role in cancer, Endocrine Related Cancer, vol.21, issue.5, pp.345-356, 2014.
DOI : 10.1530/ERC-14-0141

URL : http://erc.endocrinology-journals.org/content/21/5/R345.full.pdf

V. Luga, L. Zhang, A. Viloria-petit, A. Ogunjimi, M. Inanlou et al., Exosomes Mediate Stromal Mobilization of Autocrine Wnt-PCP Signaling in Breast Cancer Cell Migration, Cell, vol.151, issue.7, pp.1542-56, 2012.
DOI : 10.1016/j.cell.2012.11.024

R. Yagyu, R. Hamamoto, Y. Furukawa, H. Okabe, T. Yamamura et al., Isolation and characterization of a novel human gene, VANGL1, as a therapeutic target for hepatocellular carcinoma, International Journal of Oncology, 2002.
DOI : 10.3892/ijo.20.6.1173

J. Lee, J. Bae, E. Sun, H. Kim, T. Yoon et al., KITENIN increases invasion and migration of mouse squamous cancer cells and promotes pulmonary metastasis in a mouse squamous tumor model, FEBS Letters, vol.3, issue.4, pp.711-718, 2009.
DOI : 10.1038/nrc1209

H. Ryu, Y. Park, S. Park, J. Lee, S. Cho et al., KITENIN is associated with tumor progression in human gastric cancer, Anticancer Res, vol.30, issue.9, pp.3479-86, 2010.

J. Hwang, H. Shim, Y. Park, S. Cho, W. Bae et al., Intravenous KITENIN shRNA Injection Suppresses Hepatic Metastasis and Recurrence of Colon Cancer in an Orthotopic Mouse Model, Journal of Korean Medical Science, vol.26, issue.11, pp.1439-1484, 2011.
DOI : 10.3346/jkms.2011.26.11.1439

T. Yoon, S. Kim, J. Lee, Y. Park, G. Kim et al., Expression of KITENIN and its association with tumor progression in oral squamous cell carcinoma, Auris Nasus Larynx, vol.40, issue.2, 2013.
DOI : 10.1016/j.anl.2012.07.006

J. Lee, S. Park, K. Chay, Y. Seo, H. Kook et al., KAI1 COOH-Terminal Interacting Tetraspanin (KITENIN), a Member of the Tetraspanin Family, Interacts with KAI1, a Tumor Metastasis Suppressor, and Enhances Metastasis of Cancer, Cancer Research, vol.64, issue.12, pp.4235-4278, 2004.
DOI : 10.1158/0008-5472.CAN-04-0275

M. Kaucká, K. Plevová, S. Pavlová, P. Janovská, A. Mishra et al., The Planar Cell Polarity Pathway Drives Pathogenesis of Chronic Lymphocytic Leukemia by the Regulation of B-Lymphocyte Migration, Cancer Research, vol.73, issue.5, pp.1491-501, 2013.
DOI : 10.1158/0008-5472.CAN-12-1752

L. Mcshane, D. Altman, W. Sauerbrei, S. Taube, M. Gion et al., REporting recommendations for tumour MARKer prognostic studies (REMARK), European Journal of Cancer, vol.41, issue.12, pp.387-91, 2005.
DOI : 10.1016/j.ejca.2005.03.032

J. Fergenbaum, M. Garcia-closas, S. Hewitt, J. Lissowska, L. Sakoda et al., Loss of antigenicity in stored sections of breast cancer tissue microarrays. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol, pp.667-72, 2004.

L. Van-den-broek and M. Van-de-vijver, Assessment of problems in diagnostic and research immunohistochemistry associated with epitope instability in stored paraffin sections, Appl Immunohistochem Mol Morphol AIMM, vol.8, issue.4, pp.316-337, 2000.

T. Kivelä and P. Grambsch, Evaluation of Sampling Strategies for Modeling Survival of Uveal Malignant Melanoma, Investigative Opthalmology & Visual Science, vol.44, issue.8, pp.3288-93, 2003.
DOI : 10.1167/iovs.02-1328

C. Hudis, W. Barlow, J. Costantino, R. Gray, K. Pritchard et al., Proposal for Standardized Definitions for Efficacy End Points in Adjuvant Breast Cancer Trials: The STEEP System, Journal of Clinical Oncology, vol.25, issue.15, pp.2127-2159, 2007.
DOI : 10.1200/JCO.2006.10.3523

J. Beyersmann, M. Wolkewitz, and M. Schumacher, The impact of time-dependent bias in proportional hazards modelling, Statistics in Medicine, vol.5, issue.5, pp.6439-54, 2008.
DOI : 10.1007/978-1-4757-3294-8

C. Van-walraven, D. Davis, A. Forster, and G. Wells, Time-dependent bias was common in survival analyses published in leading clinical journals, Journal of Clinical Epidemiology, vol.57, issue.7, pp.672-82, 2004.
DOI : 10.1016/j.jclinepi.2003.12.008

D. Moher, K. Schulz, D. Altman, and C. Group, The CONSORT Statement: revised recommendations for improving the quality of reports of parallel-group randomized trials, Explore N Y N, 2001.

E. Von-elm, D. Altman, M. Egger, S. Pocock, P. Gøtzsche et al., The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) Statement, Epidemiology, vol.18, issue.6, pp.1495-1504, 2014.
DOI : 10.1097/EDE.0b013e3181577654

S. Mallett, A. Timmer, W. Sauerbrei, and D. Altman, Reporting of prognostic studies of tumour markers: a review of published articles in relation to REMARK guidelines, British Journal of Cancer, vol.40, issue.1, pp.173-80, 2009.
DOI : 10.3310/hta13050

D. Hayes, R. Bast, C. Desch, H. Fritsche, N. Kemeny et al., Tumor Marker Utility Grading System: a Framework to Evaluate Clinical Utility of Tumor Markers, JNCI Journal of the National Cancer Institute, vol.88, issue.20, pp.1456-66, 1996.
DOI : 10.1093/jnci/88.20.1456

D. Hayes, B. Trock, and A. Harris, Assessing the clinical impact of prognostic factors: when is " statistically significant " clinically useful?, Breast Cancer Res Treat, vol.52, pp.1-3305, 1998.

R. Simon, S. Paik, and D. Hayes, Use of Archived Specimens in Evaluation of Prognostic and Predictive Biomarkers, JNCI Journal of the National Cancer Institute, vol.101, issue.21, pp.1446-52, 2009.
DOI : 10.1093/jnci/djp335

E. Luporsi, J. Bellocq, J. Barrière, J. Bonastre, J. Chetritt et al., Prognosis and predictive values for clinical utility in breast cancer management], Oncotype DX TM MammaPrint Bull Cancer, vol.1102, issue.9, pp.719-748, 2015.

M. Duffy, P. Mcgowan, N. Harbeck, C. Thomssen, and M. Schmitt, uPA and PAI-1 as biomarkers in breast cancer: validated for clinical use in level-of-evidence-1 studies, Breast Cancer Research, vol.108, issue.Suppl, p.428, 2014.
DOI : 10.1038/bjc.2013.62

E. Luporsi, F. André, F. Spyratos, P. Martin, J. Jacquemier et al., Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review, Breast Cancer Research and Treatment, vol.63, issue.Suppl, pp.895-915
DOI : 10.1136/jcp.2010.077578

S. Mook, H. Bonnefoi, G. Pruneri, D. Larsimont, J. Jaskiewicz et al., Daily clinical practice of fresh tumour tissue freezing and gene expression profiling; logistics pilot study preceding the MINDACT trial, European Journal of Cancer, vol.45, issue.7, pp.1201-1209, 1990.
DOI : 10.1016/j.ejca.2009.01.004

F. Cardoso, L. Van-'t-veer, E. Rutgers, S. Loi, S. Mook et al., Clinical Application of the 70-Gene Profile: The MINDACT Trial, Journal of Clinical Oncology, vol.26, issue.5, pp.729-764, 2008.
DOI : 10.1200/JCO.2007.14.3222