, Le cancer du sein -les cancers les plus fréquents Disponible sur: http://www.e-cancer.fr/Professionnels-de-sante/Les- chiffres-du-cancer-en-France/Epidemiologie-des-cancers/Les-cancers-les-plus-frequents/Cancer- du-sein, Institut National du Cancer, 2018.

J. Kelsey, M. Gammon, and E. John, Reproductive Factors and Breast Cancer, Epidemiologic Reviews, vol.15, issue.1, pp.36-47, 1993.
DOI : 10.1093/oxfordjournals.epirev.a036115

K. Mcpherson, C. Steel, and J. Dixon, ABC of breast diseases: Breast cancer---epidemiology, risk factors, and genetics, BMJ, vol.321, issue.7261, pp.624-632, 2000.
DOI : 10.1136/bmj.321.7261.624

W. Dupont and D. Page, Risk Factors for Breast Cancer in Women with Proliferative Breast Disease, New England Journal of Medicine, vol.312, issue.3, pp.146-51, 1985.
DOI : 10.1056/NEJM198501173120303

H. Autorité and D. Santé, Institu National du Cancer. Guide-Affection longue durée. Tumeur maligne, affection maligne du tissu lymphatique ou hématopoïétique : cancer du sein, 2010.

A. Wolff, M. Hammond, D. Hicks, M. Dowsett, L. Mcshane et al., Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update, Journal of Clinical Oncology, vol.31, issue.31, pp.3997-4013, 2013.
DOI : 10.1200/JCO.2013.50.9984

W. Foulkes, I. Smith, and J. Reis-filho, Triple-Negative Breast Cancer, New England Journal of Medicine, vol.363, issue.20, 2010.
DOI : 10.1056/NEJMra1001389

K. Bauer, M. Brown, R. Cress, C. Parise, and V. Caggiano, Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype, Cancer, vol.12, issue.9, pp.1721-1729, 2007.
DOI : 10.1093/jnci/94.7.490

L. Carey, C. Perou, C. Livasy, L. Dressler, D. Cowan et al., Race, Breast Cancer Subtypes, and Survival in the Carolina Breast Cancer Study, JAMA, vol.295, issue.21, pp.2492-502, 2006.
DOI : 10.1001/jama.295.21.2492

R. Millikan, B. Newman, C. Tse, P. Moorman, K. Conway et al., Epidemiology of basal-like breast cancer, Breast Cancer Research and Treatment, vol.315, issue.1 Suppl, pp.123-162, 2008.
DOI : 10.1093/jnci/djh025

R. Atkinson, R. El-zein, V. Valero, A. Lucci, T. Bevers et al., Epidemiological risk factors associated with inflammatory breast cancer subtypes, Cancer Causes & Control, vol.307, issue.6, pp.359-66, 2016.
DOI : 10.1001/jama.2012.39

R. Dent, M. Trudeau, K. Pritchard, W. Hanna, H. Kahn et al., Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence, Clinical Cancer Research, vol.13, issue.15, pp.4429-4463, 2007.
DOI : 10.1158/1078-0432.CCR-06-3045

K. Collett, I. Stefansson, J. Eide, A. Braaten, H. Wang et al., A Basal Epithelial Phenotype Is More Frequent in Interval Breast Cancers Compared with Screen Detected Tumors, Cancer Epidemiology Biomarkers & Prevention, vol.14, issue.5, pp.1108-1120, 2005.
DOI : 10.1158/1055-9965.EPI-04-0394

W. Yang, M. Dryden, K. Broglio, M. Gilcrease, S. Dawood et al., Mammographic features of triple receptor-negative primary breast cancers in young premenopausal women, Breast Cancer Research and Treatment, vol.61, issue.3, pp.405-415, 2008.
DOI : 10.1259/bjr.73.871.11089459

S. Basu, W. Chen, J. Tchou, A. Mavi, T. Cermik et al., Comparison of triple-negative and estrogen receptor-positive/progesterone receptor-positive/HER2-negative breast carcinoma using quantitative fluorine-18 fluorodeoxyglucose/positron emission tomography imaging parameters, Cancer, vol.47, issue.5, pp.995-1000, 2008.
DOI : 10.1002/cncr.23226

M. Cheang, D. Voduc, C. Bajdik, S. Leung, S. Mckinney et al., Basal-Like Breast Cancer Defined by Five Biomarkers Has Superior Prognostic Value than Triple-Negative Phenotype, Clinical Cancer Research, vol.14, issue.5, pp.1368-76, 2008.
DOI : 10.1158/1078-0432.CCR-07-1658

P. Boyle, Triple-negative breast cancer: epidemiological considerations and recommendations, Annals of Oncology, vol.15, issue.8, pp.7-12, 2012.
DOI : 10.1038/nm0809-842

URL : https://hal.archives-ouvertes.fr/hal-01262979

E. Rakha, J. Reis-filho, and I. Ellis, Basal-Like Breast Cancer: A Critical Review, Journal of Clinical Oncology, vol.26, issue.15, pp.2568-81, 2008.
DOI : 10.1200/JCO.2007.13.1748

A. Vincent-salomon, G. Macgrogan, E. Charaffe-jauffret, J. Jacquemier, and L. Arnould, Identification of basal-like carcinomas in clinical practice: « triple zero/BRCA1-like » carcinomas], Bull Cancer, vol.97, issue.3, pp.357-63, 2010.

J. Jacquemier, L. Padovani, L. Rabayrol, S. Lakhani, F. Penault-llorca et al., Typical medullary breast carcinomas have a basal/myoepithelial phenotype, The Journal of Pathology, vol.10, issue.3, pp.260-268, 2005.
DOI : 10.1097/01.LAB.0000038508.86221.B3

B. Weigelt, B. Kreike, and J. Reis-filho, Metaplastic breast carcinomas are basal-like breast cancers: a genomic profiling analysis, Breast Cancer Research and Treatment, vol.25, issue.2, pp.273-80, 2009.
DOI : 10.1007/978-3-662-30514-0_3

P. Farmer, H. Bonnefoi, V. Becette, M. Tubiana-hulin, P. Fumoleau et al., Identification of molecular apocrine breast tumours by microarray analysis, Oncogene, vol.24, issue.29, pp.4660-71, 2005.
DOI : 10.1073/pnas.201162998

URL : https://breast-cancer-research.biomedcentral.com/track/pdf/10.1186/bcr1122?site=breast-cancer-research.biomedcentral.com

J. Adélaïde, P. Finetti, I. Bekhouche, L. Repellini, J. Geneix et al., Integrated Profiling of Basal and Luminal Breast Cancers, Cancer Research, vol.67, issue.24, pp.11565-75, 2007.
DOI : 10.1158/0008-5472.CAN-07-2536

S. Shah, A. Roth, R. Goya, A. Oloumi, G. Ha et al., The clonal and mutational evolution spectrum of primary triple-negative breast cancers, Nature, vol.464, issue.7403, pp.395-404, 2012.
DOI : 10.1038/nature08989

URL : http://europepmc.org/articles/pmc3863681?pdf=render

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

N. Turner and J. Reis-filho, Basal-like breast cancer and the BRCA1 phenotype, Oncogene, vol.25, issue.43, pp.5846-53, 2006.
DOI : 10.1073/pnas.171174298

URL : http://www.nature.com/onc/journal/v25/n43/pdf/1209876a.pdf

N. Turner, J. Reis-filho, A. Russell, R. Springall, K. Ryder et al., BRCA1 dysfunction in sporadic basal-like breast cancer, Oncogene, vol.24, issue.14, pp.2126-2158, 2007.
DOI : 10.1038/sj.onc.1208538

URL : http://www.nature.com/onc/journal/v26/n14/pdf/1210014a.pdf

A. Gonzalez-angulo, K. Timms, S. Liu, H. Chen, J. Litton et al., Incidence and Outcome of BRCA Mutations in Unselected Patients with Triple Receptor-Negative Breast Cancer, Clinical Cancer Research, vol.17, issue.5, pp.1082-1091, 2011.
DOI : 10.1158/1078-0432.CCR-10-2560

URL : http://clincancerres.aacrjournals.org/content/clincanres/17/5/1082.full.pdf

G. Lewis, A. Subhawong, H. Nassar, R. Vang, P. Illei et al., Relationship Between Molecular Subtype of Invasive Breast Carcinoma and Expression of Gross Cystic Disease Fluid Protein 15 and Mammaglobin, American Journal of Clinical Pathology, vol.135, issue.4, pp.587-91, 2011.
DOI : 10.1097/PAS.0b013e31817f9790

D. Byrne, S. Deb, E. Takano, and S. Fox, GATA3 expression in triple-negative breast cancers, Histopathology, vol.6, issue.1, pp.63-71, 2017.
DOI : 10.1186/1471-2407-6-96

A. Cimino-mathews, A. Subhawong, P. Illei, R. Sharma, M. Halushka et al., GATA3 expression in breast carcinoma: utility in triple-negative, sarcomatoid, and metastatic carcinomas, Human Pathology, vol.44, issue.7, pp.1341-1350, 2013.
DOI : 10.1016/j.humpath.2012.11.003

URL : http://europepmc.org/articles/pmc3991123?pdf=render

H. Liu, J. Shi, J. Prichard, Y. Gong, and F. Lin, Immunohistochemical Evaluation of GATA-3 Expression in ER-Negative Breast Carcinomas, American Journal of Clinical Pathology, vol.25, issue.S2, pp.648-55, 2014.
DOI : 10.1309/AJCP0Q9UQTEESLHN

URL : https://academic.oup.com/ajcp/article-pdf/141/5/648/4991393/ajcpath141-0648.pdf

G. Krings, M. Nystrom, I. Mehdi, P. Vohra, and Y. Chen, Diagnostic utility and sensitivities of GATA3 antibodies in triple-negative breast cancer, Human Pathology, vol.45, issue.11, pp.2225-2257, 2014.
DOI : 10.1016/j.humpath.2014.06.022

S. Santuario-facio, S. Cardona-huerta, Y. Perez-paramo, V. Trevino, F. Hernandez-cabrera et al., A New Gene Expression Signature for Triple-Negative Breast Cancer Using Frozen Fresh Tissue before Neoadjuvant Chemotherapy, Molecular Medicine, vol.23, issue.1, 2017.
DOI : 10.2119/molmed.2016.00257

URL : https://doi.org/10.2119/molmed.2016.00257

A. Rody, T. Karn, C. Liedtke, L. Pusztai, E. Ruckhaeberle et al., A clinically relevant gene signature in triple negative and basal-like breast cancer, Breast Cancer Research, vol.54, issue.5, p.97, 2011.
DOI : 10.1002/jcb.240540406

URL : https://breast-cancer-research.biomedcentral.com/track/pdf/10.1186/bcr3035?site=breast-cancer-research.biomedcentral.com

M. Komatsu, T. Yoshimaru, T. Matsuo, K. Kiyotani, Y. Miyoshi et al., Molecular features of triple negative breast cancer cells by genome-wide gene expression profiling analysis, International Journal of Oncology, vol.42, issue.2, pp.478-506, 2013.
DOI : 10.3892/ijo.2012.1744

F. Al-ejeh, P. Simpson, J. Sanus, K. Klein, M. Kalimutho et al., Meta-analysis of the global gene expression profile of triple-negative breast cancer identifies genes for the prognostication and treatment of aggressive breast cancer, p.100, 2014.

L. Cascione, P. Gasparini, F. Lovat, S. Carasi, A. Pulvirenti et al., Integrated MicroRNA and mRNA Signatures Associated with Survival in Triple Negative Breast Cancer, PLoS ONE, vol.411, issue.2, p.55910, 2013.
DOI : 10.1371/journal.pone.0055910.s009

URL : https://doi.org/10.1371/journal.pone.0055910

D. Nonaka, L. Chiriboga, and B. Rubin, Sox10: A Pan-Schwannian and Melanocytic Marker, The American Journal of Surgical Pathology, vol.32, issue.9, pp.1291-1299, 2008.
DOI : 10.1097/PAS.0b013e3181658c14

R. Mollaaghababa and W. Pavan, The importance of having your SOX on: role of SOX10??? in the development of neural crest-derived melanocytes and glia, Oncogene, vol.22, issue.20, pp.3024-3058, 2003.
DOI : 10.1016/S0896-6273(00)80898-3

R. Kelsh, Sorting outSox10 functions in neural crest development, BioEssays, vol.10, issue.8, pp.788-98, 2006.
DOI : 10.1017/CBO9780511525322

Y. Zhu, Y. Jia, L. Hu, C. Qi, M. Prasad et al., Peroxisome-proliferator-activated receptor-binding protein (PBP) is essential for the growth of active Notch4-immortalized mammary epithelial cells by activating SOX10 expression, Biochemical Journal, vol.1, issue.2, pp.435-479, 2009.
DOI : 10.1128/MCB.00967-07

S. Artavanis-tsakonas, M. Rand, and R. Lake, Notch Signaling: Cell Fate Control and Signal Integration in Development, Science, vol.284, issue.5415, pp.770-776, 1999.
DOI : 10.1126/science.284.5415.770

W. Feng, S. Liu, R. Zhu, B. Li, Z. Zhu et al., SOX10 induced Nestin expression regulates cancer stem cell properties of TNBC cells, Biochemical and Biophysical Research Communications, vol.485, issue.2, pp.522-530, 2017.
DOI : 10.1016/j.bbrc.2017.02.014

S. Ivanov, A. Panaccione, D. Nonaka, M. Prasad, K. Boyd et al., Diagnostic SOX10 gene signatures in salivary adenoid cystic and breast basal-like carcinomas, British Journal of Cancer, vol.109, issue.2, pp.444-51, 2013.
DOI : 10.1074/jbc.M110.149658

URL : http://www.nature.com/bjc/journal/v109/n2/pdf/bjc2013326a.pdf

M. Miettinen, P. Mccue, M. Sarlomo-rikala, W. Biernat, P. Czapiewski et al., Sox10???A Marker for Not Only Schwannian and Melanocytic Neoplasms But Also Myoepithelial Cell Tumors of Soft Tissue, The American Journal of Surgical Pathology, vol.39, issue.6, pp.826-861, 2015.
DOI : 10.1097/PAS.0000000000000398

A. Cimino-mathews, A. Subhawong, H. Elwood, H. Warzecha, R. Sharma et al., Neural crest transcription factor Sox10 is preferentially expressed in triple-negative and metaplastic breast carcinomas, Human Pathology, vol.44, issue.6, pp.959-65, 2013.
DOI : 10.1016/j.humpath.2012.09.005

URL : http://europepmc.org/articles/pmc3978178?pdf=render

C. Dravis, B. Spike, J. Harrell, C. Johns, C. Trejo et al., Sox10 Regulates Stem/Progenitor and Mesenchymal Cell States in Mammary Epithelial Cells. Cell Rep. 29 sept, vol.12, issue.12, pp.2035-2083, 2015.

C. Riemenschnitter, I. Teleki, V. Tischler, W. Guo, and Z. Varga, Stability and prognostic value of Slug, Sox9 and Sox10 expression in breast cancers treated with neoadjuvant chemotherapy, SpringerPlus, vol.2, issue.1, p.695, 2013.
DOI : 10.1007/s00428-004-1164-4

M. Asselin-labat, K. Sutherland, H. Barker, R. Thomas, M. Shackleton et al., Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation, Nature Cell Biology, vol.98, issue.2, pp.201-210, 2007.
DOI : 10.1073/pnas.251547698

B. Rahim, O. Regan, and R. , AR Signaling in Breast Cancer, Cancers, vol.6, issue.12, 2017.
DOI : 10.1186/bcr577

M. Schweizer and E. Yu, AR-Signaling in Human Malignancies: Prostate Cancer and Beyond, Cancers, vol.11, issue.12, 2017.
DOI : 10.1002/hed.23383

R. Berardi, F. Morgese, A. Santinelli, A. Onofri, T. Biscotti et al., Hormonal receptors in lung adenocarcinoma: expression and difference in outcome by sex, Oncotarget, vol.7, issue.50, pp.82648-57, 2016.
DOI : 10.18632/oncotarget.12244

, Institut National du cancer -les chiffres du cancer en France - Epidémiologie des cancers -Les cancers les plus fréquents -le cancer du poumon, Institut National du Cancer

. France, Epidemiologie-des-cancers/Les-cancers-les-plus-frequents/Cancer-du-poumon

D. Cruz, C. Tanoue, L. Matthay, and R. , Lung Cancer: Epidemiology, Etiology, and Prevention, Clinics in Chest Medicine, vol.32, issue.4, pp.605-649, 2011.
DOI : 10.1016/j.ccm.2011.09.001

N. Howlader, A. Noone, M. Krapcho, N. Neyman, R. Aminou et al., SEER Cancer Statistics Review National Cancer Institute. Bethesda, MD [Internet]. SEER Cancer Statistics Review, 1975.

W. Foulkes, I. Smith, and J. Reis-filho, Triple-Negative Breast Cancer, New England Journal of Medicine, vol.363, issue.20, pp.1938-1986, 2010.
DOI : 10.1056/NEJMra1001389

F. Penault-llorca and N. Radosevic-robin, Biomarkers of residual disease after neoadjuvant therapy for breast cancer, Nature Reviews Clinical Oncology, vol.22, issue.8, pp.487-503, 2016.
DOI : 10.1016/j.ejca.2014.11.018

L. Esserman, D. Moore, and P. Tsing, Biologic markers determine both the risk and the timing of recurrence in breast cancer, Breast Cancer Research and Treatment, vol.55, issue.4, pp.607-623, 2011.
DOI : 10.1200/JCO.2005.06.178

P. Boyle, Triple-negative breast cancer: epidemiological considerations and recommendations, Annals of Oncology, vol.15, issue.8, pp.7-12, 2012.
DOI : 10.1038/nm0809-842

URL : https://hal.archives-ouvertes.fr/hal-01262979

R. Dent, W. Hanna, and M. Trudeau, Pattern of metastatic spread in triple-negative breast cancer, Breast Cancer Research and Treatment, vol.356, issue.2, pp.423-431, 2009.
DOI : 10.1093/jnci/djg050

H. Tanaka, H. Tsukuma, and H. Koyama, Second Primary Cancers Following Breast Cancer in the Japanese Female Population, Japanese Journal of Cancer Research, vol.56, issue.1, pp.1-8, 2001.
DOI : 10.1093/jnci/89.3.227

E. Buiatti, E. Crocetti, and S. Acciai, Incidence of second primary cancers in three Italian population-based cancer registries, European Journal of Cancer, vol.33, issue.11, pp.1829-1863, 1997.
DOI : 10.1016/S0959-8049(97)00173-1

H. Evans, C. Lewis, and D. Robinson, Incidence of multiple primary cancers in a cohort of women diagnosed with breast cancer in southeast England, British Journal of Cancer, vol.84, issue.3, pp.435-475, 2001.
DOI : 10.1054/bjoc.2000.1603

M. Schaapveld, O. Visser, and M. Louwman, Risk of New Primary Nonbreast Cancers After Breast Cancer Treatment: A Dutch Population-Based Study, Journal of Clinical Oncology, vol.26, issue.8, pp.1239-1285, 2008.
DOI : 10.1200/JCO.2007.11.9081

L. Mellemkjaer, S. Friis, and J. Olsen, Risk of second cancer among women with breast cancer, International Journal of Cancer, vol.111, issue.9, pp.2285-92, 2006.
DOI : 10.1111/j.1349-7006.2001.tb01040.x

B. Turner, P. Cagle, and I. Sainz, Napsin A, a New Marker for Lung Adenocarcinoma, Is Complementary and More Sensitive and Specific Than Thyroid Transcription Factor 1 in the Differential Diagnosis of Primary Pulmonary Carcinoma: Evaluation of 1674 Cases by Tissue Microarray, Archives of Pathology & Laboratory Medicine, vol.136, issue.2, pp.163-71, 2012.
DOI : 10.5858/arpa.2011-0320-OA

J. Bishop, R. Sharma, and P. Illei, Napsin A and thyroid transcription factor-1 expression in carcinomas of the lung, breast, pancreas, colon, kidney, thyroid, and malignant mesothelioma, Human Pathology, vol.41, issue.1, pp.20-25, 2010.
DOI : 10.1016/j.humpath.2009.06.014

B. Lehmann, J. Bauer, and X. Chen, Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies, Journal of Clinical Investigation, vol.121, issue.7, 2011.
DOI : 10.1172/JCI45014DS1

M. Burstein, A. Tsimelzon, and G. Poage, Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res, pp.1688-98, 2015.
DOI : 10.1158/1078-0432.ccr-14-0432

URL : http://clincancerres.aacrjournals.org/content/21/7/1688.full.pdf

D. Koboldt, R. Fulton, and M. Mclellan, Comprehensive molecular portraits of human breast tumours, Nature, vol.16, issue.7418, pp.61-70, 2012.
DOI : 10.1016/j.ccr.2009.06.006

Y. Yang, S. Lu, and W. Zeng, GATA3 expression in clinically useful groups of breast carcinoma: a comparison with GCDFP15 and mammaglobin for identifying paired primary and metastatic tumors, Annals of Diagnostic Pathology, vol.26, pp.20171-20176
DOI : 10.1016/j.anndiagpath.2016.09.011

L. Huo, J. Zhang, and M. Gilcrease, Gross cystic disease fluid protein-15 and mammaglobin A expression determined by immunohistochemistry is of limited utility in triple-negative breast cancer, Histopathology, vol.50, issue.2, pp.267-74, 2013.
DOI : 10.1007/BF02967653

T. Shaoxian, Y. Baohua, and X. Xiaoli, Characterisation of GATA3 expression in invasive breast cancer: differences in histological subtypes and immunohistochemically defined molecular subtypes, Journal of Clinical Pathology, vol.45, issue.11, pp.926-960, 2017.
DOI : 10.1136/jclinpath-2016-204137

R. Bhargava, S. Beriwal, and D. Dabbs, Mammaglobin vs GCDFP-15, American Journal of Clinical Pathology, vol.127, issue.1, pp.103-116, 2007.
DOI : 10.1309/TDP92PQLDE2HLEET

URL : https://academic.oup.com/ajcp/article-pdf/127/1/103/24984749/ajcpath127-0103.pdf

M. Miettinen, P. Mccue, and M. Sarlomo-rikala, GATA3, The American Journal of Surgical Pathology, vol.38, issue.1, pp.13-22, 2014.
DOI : 10.1097/PAS.0b013e3182a0218f

G. Lewis, A. Subhawong, and H. Nassar, Relationship Between Molecular Subtype of Invasive Breast Carcinoma and Expression of Gross Cystic Disease Fluid Protein 15 and Mammaglobin, American Journal of Clinical Pathology, vol.135, issue.4, pp.587-91, 2011.
DOI : 10.1097/PAS.0b013e31817f9790

D. Byrne, S. Deb, and E. Takano, GATA3 expression in triple-negative breast cancers, Histopathology, vol.6, issue.1, pp.63-71
DOI : 10.1186/1471-2407-6-96

A. Cimino-mathews, A. Subhawong, and P. Illei, GATA3 expression in breast carcinoma: utility in triple-negative, sarcomatoid, and metastatic carcinomas, Human Pathology, vol.44, issue.7, 2013.
DOI : 10.1016/j.humpath.2012.11.003

H. Liu, J. Shi, and J. Prichard, Immunohistochemical Evaluation of GATA-3 Expression in ER-Negative Breast Carcinomas, American Journal of Clinical Pathology, vol.25, issue.S2, pp.648-55, 2014.
DOI : 10.1309/AJCP0Q9UQTEESLHN

G. Krings, M. Nystrom, and I. Mehdi, Diagnostic utility and sensitivities of GATA3 antibodies in triple-negative breast cancer, Human Pathology, vol.45, issue.11, pp.2225-2257, 2014.
DOI : 10.1016/j.humpath.2014.06.022

A. Mina, R. Yoder, and P. Sharma, Targeting the androgen receptor in triple-negative breast cancer: current perspectives, OncoTargets and Therapy, vol.10, pp.4675-85, 2017.
DOI : 10.2147/OTT.S126051

A. Panaccione, Y. Guo, and W. Yarbrough, Expression Profiling of Clinical Specimens Supports the Existence of Neural Progenitor-Like Stem Cells in Basal Breast Cancers. Clin Breast Cancer, pp.298-306, 2017.

P. Farmer, H. Bonnefoi, and V. Becette, Identification of molecular apocrine breast tumours by microarray analysis. Oncogene, Jul, vol.724, issue.29, pp.4660-71, 2005.

A. Wolff, M. Hammond, and D. Hicks, Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update, Journal of Clinical Oncology, vol.31, issue.31, pp.3997-4013, 2013.
DOI : 10.1200/JCO.2013.50.9984

L. Kinsel, E. Szabo, and G. Greene, Immunocytochemical analysis of estrogen receptors as a predictor of prognosis in breast cancer patients: comparison with quantitative biochemical methods. Cancer Res, Feb, vol.1549, issue.4, pp.1052-1058, 1989.

W. Travis, E. Brambilla, and A. Burke, WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart. WHO Classification of Tumours, International Agency for Research on Cancer, vol.7, 2015.

J. Brierley, M. Gospodarowicz, and C. Wittekind, TNM Classification of Malignant Tumors, Eighth edition, 2017.

M. Prochazka, P. Hall, and G. Gagliardi, Ionizing Radiation and Tobacco Use Increases the Risk of a Subsequent Lung Carcinoma in Women With Breast Cancer: Case-Only Design, Journal of Clinical Oncology, vol.23, issue.30, pp.7467-74, 2005.
DOI : 10.1200/JCO.2005.01.7335

S. Schonfeld, R. Curtis, and W. Anderson, The risk of a second primary lung cancer after a first invasive breast cancer according to estrogen receptor status, Cancer Causes & Control, vol.171, issue.2, pp.1721-1729, 2012.
DOI : 10.1001/archinternmed.2010.503

P. Inskip, M. Stovall, and J. Flannery, Lung Cancer Risk and Radiation Dose Among Women Treated for Breast Cancer, JNCI Journal of the National Cancer Institute, vol.86, issue.13, pp.983-991, 1994.
DOI : 10.1093/jnci/86.13.983

T. Grantzau and J. Overgaard, Risk of second non-breast cancer among patients treated with and without postoperative radiotherapy for primary breast cancer: A systematic review and meta-analysis of population-based studies including 522,739 patients, Radiotherapy and Oncology, vol.121, issue.3, 2016.
DOI : 10.1016/j.radonc.2016.08.017

L. Bazire, Y. De-rycke, and B. Asselain, Risks of second malignancies after breast cancer treatment: Long-term results. Cancer/Radiothérapie, Feb, vol.21, issue.1, pp.10-15, 2017.

N. P. Wang, S. Zee, and R. Zarbo, Coordinate Expression of Cytokeratins 7 and 20 Defines Unique Subsets of Carcinomas. Appl Immunohistochem, Jul, vol.13, issue.2, pp.99-107, 1995.

C. Perou, T. Sørlie, and M. Eisen, Molecular portraits of human breast tumours, Nature, vol.278, issue.6797, pp.747-52, 2000.
DOI : 10.1038/35000501

F. Bertucci, P. Finetti, and N. Cervera, How basal are triple-negative breast cancers? Int J Cancer, pp.236-276, 2008.

A. Rody, T. Karn, and C. Liedtke, A clinically relevant gene signature in triple negative and basal-like breast cancer, Breast Cancer Research, vol.54, issue.5, p.97, 2011.
DOI : 10.1002/jcb.240540406

J. Choo and T. Nielsen, Biomarkers for Basal-like Breast Cancer, Cancers, vol.119, issue.2, pp.1040-65, 2010.
DOI : 10.1158/1078-0432.CCR-08-1208

T. Nielsen, F. Hsu, and K. Jensen, Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res, Aug, vol.1510, issue.16, pp.5367-74, 2004.

C. Livasy, G. Karaca, and R. Nanda, Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol, Feb, vol.19, issue.2, pp.264-71, 2006.

J. Won, D. Gao, and C. Chow, A survey of immunohistochemical biomarkers for basal-like breast cancer against a gene expression profile gold standard. Mod Pathol, 2013.

D. Laurence, P. Monaghan, and B. Gusterson, The development of the normal human breast, Oxf Rev Reprod Biol, vol.13, pp.149-74, 1991.

L. Reid, The Embryology of the Lung, Ciba Foundation Symposium : Development of the Lung, pp.109-139, 1967.
DOI : 10.1002/9780470719473.ch7

R. Mollaaghababa and W. Pavan, The importance of having your SOX on: role of SOX10??? in the development of neural crest-derived melanocytes and glia, Oncogene, vol.22, issue.20, pp.3024-3058, 2003.
DOI : 10.1016/S0896-6273(00)80898-3

R. Kelsh, Sorting out Sox10 functions in neural crest development, BioEssays, 2006.

Y. Zhu, Y. Jia, and L. Hu, Peroxisome-proliferator-activated receptor-binding protein (PBP) is essential for the growth of active Notch4-immortalized mammary epithelial cells by activating SOX10 expression, Biochemical Journal, vol.1, issue.2, pp.435-479, 2009.
DOI : 10.1128/MCB.00967-07

W. Feng, S. Liu, and R. Zhu, SOX10 induced Nestin expression regulates cancer stem cell properties of TNBC cells, Biochemical and Biophysical Research Communications, vol.485, issue.2, pp.522-530
DOI : 10.1016/j.bbrc.2017.02.014

D. Nonaka, L. Chiriboga, and B. Rubin, Sox10: A Pan-Schwannian and Melanocytic Marker, The American Journal of Surgical Pathology, vol.32, issue.9, pp.1291-1299, 2008.
DOI : 10.1097/PAS.0b013e3181658c14

A. Cimino-mathews, A. Subhawong, and H. Elwood, Neural crest transcription factor Sox10 is preferentially expressed in triple-negative and metaplastic breast carcinomas, Human Pathology, vol.44, issue.6, p.2013
DOI : 10.1016/j.humpath.2012.09.005

M. Miettinen, P. Mccue, and M. Sarlomo-rikala, Sox10???A Marker for Not Only Schwannian and Melanocytic Neoplasms But Also Myoepithelial Cell Tumors of Soft Tissue, The American Journal of Surgical Pathology, vol.39, issue.6, pp.826-861
DOI : 10.1097/PAS.0000000000000398

E. Nelson, R. Sharma, and P. Argani, Utility of Sox10 labeling in metastatic breast carcinomas, Human Pathology, vol.67, pp.205-215
DOI : 10.1016/j.humpath.2017.08.011

M. Asselin-labat, K. Sutherland, and H. Barker, Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation, Nature Cell Biology, vol.98, issue.2, pp.201-210, 2007.
DOI : 10.1073/pnas.251547698

B. Rahim, O. Regan, and R. , AR Signaling in Breast Cancer, Cancers, vol.6, issue.12
DOI : 10.1186/bcr577

URL : https://www.mdpi.com/2072-6694/9/3/21/pdf

M. Schweizer and E. Yu, AR-Signaling in Human Malignancies: Prostate Cancer and Beyond, Cancers, vol.11, issue.12, 2017.
DOI : 10.1002/hed.23383

R. Berardi, F. Morgese, and A. Santinelli, Hormonal receptors in lung adenocarcinoma: expression and difference in outcome by sex, Oncotarget, vol.7, issue.50, pp.82648-57, 2016.
DOI : 10.18632/oncotarget.12244

A. Thike, Y. Chong, L. Cheok, and P. , Loss of androgen receptor expression predicts early recurrence in triple-negative and basal-like breast cancer, Modern Pathology, vol.55, issue.3, pp.352-60, 2014.
DOI : 10.1507/endocrj.K07E-053

E. Rakha, M. Sayed, and A. Green, Prognostic markers in triple-negative breast cancer, Cancer, vol.91, issue.1, pp.25-32, 2007.
DOI : 10.1016/S0002-9440(10)64476-8

L. Niemeier, D. Dabbs, and S. Beriwal, Androgen receptor in breast cancer: expression in estrogen receptor-positive tumors and in estrogen receptor-negative tumors with apocrine differentiation, Modern Pathology, vol.185, issue.2, pp.205-217, 2010.
DOI : 10.1016/S0002-9610(03)00054-0

S. Park, K. J. Park, and H. , Expression of androgen receptors in primary breast cancer, Annals of Oncology, vol.25, issue.28, pp.488-92, 2010.
DOI : 10.1038/sj.onc.1209415

A. Peters, G. Buchanan, and C. Ricciardelli, Androgen Receptor Inhibits Estrogen Receptor-?? Activity and Is Prognostic in Breast Cancer, Cancer Research, vol.69, issue.15, pp.6131-6171, 2009.
DOI : 10.1158/0008-5472.CAN-09-0452

J. Mossler, T. Barton, and A. Brinkhous, Apocrine differentiation in human mammary carcinoma. Cancer, pp.2463-71, 1980.

P. Ray, J. Wang, Y. Qu, M. Sim, J. Shamonki et al., FOXC1 Is a Potential Prognostic Biomarker with Functional Significance in Basal-like Breast Cancer, Cancer Research, vol.70, issue.10, pp.3870-3876, 2010.
DOI : 10.1158/0008-5472.CAN-09-4120

J. Won, D. Gao, C. Chow, J. Cheng, S. Lau et al., A survey of immunohistochemical biomarkers for basal-like breast cancer against a gene expression profile gold standard, Modern Pathology, vol.52, issue.11, pp.1438-50, 2013.
DOI : 10.1093/jnci/djn326