W. D. Foulkes, I. E. Smith, and J. S. Reis-filho, Triple-negative breast cancer, N Engl J Med, vol.363, pp.1938-1986, 2010.

S. G. Ahn, S. J. Kim, C. Kim, and J. Jeong, Molecular classification of triple-negative breast cancer, J Breast Cancer, vol.19, pp.223-253, 2016.

H. Bonsang-kitzis, B. Sadacca, A. S. Hamy-petit, M. Moarii, A. Pinheiro et al., Biological network-driven gene selection identifies a stromal immune module as a key determinant of triple-negative breast carcinoma prognosis, Oncoimmunology, vol.5, p.1061176, 2016.

B. D. Lehmann, J. A. Bauer, X. Chen, M. E. Sanders, A. B. Chakravarthy et al., Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies, J Clin Invest, vol.121, pp.2750-67, 2011.

M. D. Burstein, A. Tsimelzon, G. M. Poage, K. R. Covington, A. Contreras et al., Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer, Clin Cancer Res, vol.21, pp.1688-98, 2015.
DOI : 10.1158/1078-0432.ccr-14-0432

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

P. Jézéquel, D. Loussouarn, C. Guérin-charbonnel, L. Campion, A. Vanier et al., Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response, Breast Cancer Res, vol.17, p.43, 2015.

S. Guiu, C. Charon-barra, D. Vernerey, P. Fumoleau, M. Campone et al., Coexpression of androgen receptor and FOXA1 in nonmetastatic triple-negative breast cancer: ancillary study from PAVS08 trial, Future Oncol, vol.11, pp.2283-97, 2015.

R. Edgar, M. Domrachev, and A. E. Lash, Gene Expression Omnibus: NCBI gene expression and hybridization array data repository, Nucleic Acids Res, vol.30, pp.207-217, 2002.
DOI : 10.1093/nar/30.1.207

URL : https://academic.oup.com/nar/article-pdf/30/1/207/9901036/300207.pdf

J. Hubble, J. Demeter, J. H. Mao, M. Nitzberg, M. Reddy et al., Implementation of GenePattern within the Stanford Microarray Database, Nucleic Acids Res, vol.37, pp.898-901, 2009.

A. M. Newman, C. L. Liu, M. R. Green, A. J. Gentles, W. Feng et al., Robust enumeration of cell subsets from tissue expression profiles, Nat Methods, vol.12, pp.453-460, 2015.

J. Chen, E. E. Bardes, B. J. Aronow, and A. G. Jegga, ToppGene Suite for gene list enrichment analysis and candidate gene prioritization, Nucleic Acids Res, vol.37, pp.305-316, 2009.
DOI : 10.1093/nar/gkp427

URL : https://academic.oup.com/nar/article-pdf/37/suppl_2/W305/3956724/gkp427.pdf

F. P. O'connell, J. L. Pinkus, and G. S. Pinkus, CD138 (syndecan-1), a plasma cell marker immunohistochemical profile in hematopoietic and nonhematopoietic neoplasms, Am J Clin Pathol, vol.121, pp.254-63, 2004.

R. Salgado, C. Denkert, S. Demaria, N. Sirtaine, F. Klauschen et al., International TILs Working Group 2014: the evaluation of tumor-infiltrating (TILs) in breast cancer: recommendations by an international TILs working group, Ann Oncol, vol.26, pp.259-71, 2014.

M. L. Troxell, E. J. Schwartz, M. Van-de-rijn, D. T. Ross, R. A. Warnke et al., Follicular dendritic cell immunohistochemical markers in angioimmunoblastic T-cell lymphoma, Appl Immunohistochem Mol Morphol, vol.13, pp.297-303, 2005.
DOI : 10.1097/01.pai.0000173053.45296.51

L. Martinet, I. Garrido, T. Filleron, L. Guellec, S. Bellard et al., Human solid tumors contain high endothelial venules: association with Tand B-lymphocytes and favorable prognosis in breast cancer, Cancer Res, vol.71, pp.5678-87, 2011.
DOI : 10.1158/0008-5472.can-11-0431

URL : http://cancerres.aacrjournals.org/content/71/17/5678.full.pdf

. R-core-team, R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing, 2016.

B. D. Lehmann, J. A. Bauer, J. M. Schafer, C. S. Pendleton, L. Tang et al., PIK3CA mutations in androgen receptor-positive triple negative breast cancer confer sensitivity to the combination of PI3K and androgen receptor inhibitors, Breast Cancer Res, vol.16, p.406, 2014.

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, pp.4660-71, 2005.
DOI : 10.1186/bcr1122

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

A. S. Doane, M. Danso, P. Lal, M. Donaton, L. Zhang et al., An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen, Oncogene, vol.25, pp.3994-4008, 2006.
DOI : 10.1038/sj.onc.1209415

URL : https://www.nature.com/articles/1209415.pdf

A. Prat, B. Adamo, M. C. Cheang, C. K. Anders, L. A. Carey et al., Molecular characterization of basal-like and non-basal-like triple-negative breast cancer, Oncologist, vol.18, pp.123-156, 2013.
DOI : 10.1634/theoncologist.2012-0397

URL : http://theoncologist.alphamedpress.org/content/18/2/123.full.pdf

A. Daemen and G. Manning, HER2 is not a cancer subtype but rather a pancancer event and is highly enriched in AR-driven breast tumors, Breast Cancer Res, vol.20, p.8, 2018.

R. A. Flavell, S. Sanjabi, S. H. Wrzesinski, and P. Licona-limón, The polarization of immune cells in tumour environment by TGFbeta, Nat Rev Immunol, vol.10, pp.554-67, 2010.

A. J. Gentles, A. M. Newman, C. L. Liu, S. V. Bratman, W. Feng et al., The prognostic landscape of genes and infiltrating immune cells across human cancers, Nat Med, vol.21, pp.938-983, 2015.

A. Mantovani, A. Sica, and . Macrophages, innate immunity and cancer: balance, tolerance, and diversity, Curr Opin Immunol, vol.22, pp.231-238, 2010.
DOI : 10.1016/j.coi.2010.01.009

S. V. Torti and F. M. Torti, Iron and cancer: more ore to be miner, Nat Rev Cancer, vol.13, pp.342-55, 2013.

R. J. Deberardinis and N. S. Chandel, Fundamentals of cancer metabolism, Sci Adv, vol.2, p.1600200, 2016.

S. Deborde, T. Omelchenko, A. Lyubchik, Y. Zhou, S. He et al., Schwann cells induce cancer cell dispersion and invasion, J Clin Invest, vol.126, pp.1538-54, 2016.
DOI : 10.1172/jci82658

URL : http://www.jci.org/articles/view/82658/files/pdf

M. P. Clements, E. Byrne, C. Guerrero, L. F. Cattin, A. L. Zakka et al., The wound microenvironment reprograms Schwann cells to invasive mesenchymal-like cells to drive peripheral nerve regeneration, Neuron, vol.96, pp.98-114, 2017.

A. Kassambara, T. Rème, J. M. Fest, T. Hose, D. Tarte et al., GenomicScape: an easy-to-use web tool for gene expression data analysis. Application to investigate the molecular events in the differentiation of B cells into plasma cells, PLoS Comput Biol, vol.11, p.1004077, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01112225

F. Aloisi and R. Pujol-borrell, Lymphoid neogenesis in chronic inflammatory diseases, Nat Rev Immunol, vol.6, pp.205-222, 2006.

N. Hiraoka, Y. Ino, and R. Yamazaki-itoh, Tertiary lymphoid organs in cancer tissues, Front Immunol, vol.7, p.244, 2016.

D. Bedognetti, M. Hendrickx, and L. D. Miller, Prognostic and predictive immune gene signatures in breast cancer, Curr Opin Oncol, vol.27, pp.433-477, 2015.

Z. Q. Wang, K. Milne, H. Derocher, J. R. Webb, B. H. Nelson et al., PD-L1 and intratumoral immune response in breast cancer, Oncotarget, vol.8, pp.51641-51, 2017.

D. Simone, M. Arrigoni, A. Rossetti, G. Gruarin, P. Ranzani et al., Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells, Immunity, vol.45, pp.1135-1182, 2016.

G. Bianchini, J. M. Balko, I. A. Mayer, M. E. Sanders, and L. Gianni, Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease, Nat Rev Clin Oncol, vol.13, pp.674-90, 2016.

G. C. Monnot and P. Romero, Rationale for immunological approaches to breast cancer therapy, Breast, vol.37, pp.187-95, 2018.

D. Caro, G. Castino, G. F. Bergomas, F. , C. N. Chiriva-internati et al., Tertiary lymphoid tissue in the tumor microenvironment: from its occurrence to immunotherapeutic implications, Int Rev Immunol, vol.34, pp.123-156, 2015.

D. M. Pardoll, The blockade of immune checkpoints in cancer immunotherapy, Nat Rev Cancer, vol.12, pp.252-64, 2012.

H. L. Macgregor and P. S. Ohashi, Molecular pathways: evaluating the potential for B7-H4 as an immunoregulatory target, Clin Cancer Res, vol.23, pp.2934-2975, 2017.

I. Kryczek, L. Zou, P. Rodriguez, G. Zhu, S. Wei et al., B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma, J Exp Med, vol.203, pp.871-81, 2006.

Y. Yao, H. Ye, Z. Qi, L. Mo, Q. Yue et al., B7-H4(B7x)-mediated cross-talk between glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway lead to poor prognosis in glioma patients, Clin Cancer Res, vol.22, pp.2778-90, 2016.

D. Palma, M. Lewis, and C. E. , Macrophage regulation of tumor responses to anticancer therapies, Cancer Cell, vol.23, pp.277-86, 2013.

V. Bronte and P. J. Murray, Understanding local macrophage phenotypes in disease: modulating macrophage function to treat cancer, Nat Med, vol.21, pp.117-126, 2015.
DOI : 10.1038/nm.3794

A. Mantovani, F. Marchesi, A. Malesci, L. Laghi, and P. Allavena, Tumourassociated macrophages as treatment targets in oncology, Nat Rev Clin Oncol, vol.14, pp.399-416, 2017.

J. L. Guerriero, A. Sotayo, H. E. Ponichtera, J. A. Castrillon, A. L. Pourzia et al., Class IIa HDAC inhibition reduces breast tumours and metastases through anti-tumour macrophages, Nature, vol.542, pp.428-460, 2017.

B. Boilly, S. Faulkner, P. Jobling, and H. Hondermarck, Nerve dependence: from regeneration to cancer, Cancer Cell, vol.3, pp.342-54, 2017.

A. Vaishnavi, A. T. Le, and R. C. Doebele, TRKing down an old oncogene in a new era of targeted therapy, Cancer Discov, vol.5, pp.25-34, 2015.

P. Jobling, J. Pundavela, S. M. Oliveira, S. Roselli, M. M. Walker et al., Nerve-cancer cell cross-talk: a novel promoter of tumor progression, Cancer Res, vol.75, pp.1777-81, 2015.

C. Magnon, S. J. Hall, J. Lin, X. Xue, L. Gerber et al., Autonomic nerve development contributes to prostate progression, Science, vol.341, p.1236361, 2013.

C. M. Zhao, Y. Hayakawa, Y. Kodama, S. Muthupalani, C. B. Westphalen et al., Denervation suppresses gastric tumorigenesis, Sci Transl Med, vol.6, pp.250-115, 2014.

S. C. Peterson, M. Eberl, A. N. Vagnozzi, A. Belkadi, N. A. Veniaminova et al., Basal cell carcinoma preferentially arises from stem cells within hair follicle and mechanosensory niches, Cell Stem Cell, vol.16, pp.400-412, 2015.

J. L. Saloman, K. M. Albers, D. Li, D. J. Hartman, H. C. Crawford et al., Ablation of sensory neurons in a genetic model of pancreatic ductal adenocarcinoma slows initiation and progression of cancer, Proc Natl Acad Sci U S A, vol.113, pp.3078-83, 2016.

A. H. Zahalka, A. Arnal-estape, M. Maryanovich, F. Nakahara, C. D. Cruz et al., Adrenergic nerves activate an angio-metabolic switch in prostate cancer, Science, vol.358, pp.321-327, 2017.

J. Pundavela, S. Roselli, S. Faulkner, J. Attia, R. J. Scott et al., Nerve fibers infiltrate the tumor microenvironment and are associated with nerve growth factor production and lymph node invasion in breast cancer, Mol Oncol, vol.9, pp.1626-1661, 2015.