F. Jacques, S. Isabelle, D. Rajesh, E. Sultan, M. Colin et al., Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012, Int J Cancer, vol.136, issue.5, pp.359-386, 2014.

M. Wong, W. B. Goggins, H. Wang, F. Fung, C. Leung et al., Global Incidence and Mortality for Prostate Cancer: Analysis of Temporal Patterns and Trends in 36 Countries, Eur Urol, vol.70, issue.5, pp.862-74, 2016.

K. M. Wadosky and S. Koochekpour, Molecular mechanisms underlying resistance to androgen deprivation therapy in prostate cancer, Oncotarget, vol.7, issue.39, pp.64447-70, 2016.

P. A. Watson, V. K. Arora, and C. L. Sawyers, Emerging Mechanisms of Resistance to Androgen Receptor Inhibitors in Prostate Cancer, Nat Rev Cancer, vol.15, issue.12, pp.701-712, 2015.

E. Antonarakis, A. Armstrong, S. Dehm, and J. Luo, Androgen receptor variant-driven prostate cancer: clinical implications and therapeutic targeting. Prostate Cancer Prostatic Dis, vol.19, pp.231-272, 2016.

C. Sprenger and S. R. Plymate, The link between androgen receptor splice variants and castration resistant prostate cancer. Horm Cancer, vol.5, pp.207-224, 2014.

S. Cao, Y. Zhan, and Y. Dong, Emerging data on androgen receptor splice variants in prostate cancer, Endocr Relat Cancer, vol.23, issue.12, pp.199-210, 2016.

A. A. Shafi, A. E. Yen, and N. L. Weigel, Androgen receptors in hormone-dependent and castrationresistant prostate cancer, Pharmacol Ther, vol.140, issue.3, pp.223-261, 2013.

J. Luo, G. Attard, S. P. Balk, C. Bevan, K. Burnstein et al., Role of Androgen Receptor Variants in Prostate Cancer: Report from the 2017 Mission Androgen Receptor Variants Meeting, Eur Urol, vol.73, issue.5, pp.715-738, 2018.

K. E. Ware, M. A. Garcia-blanco, A. J. Armstrong, and S. M. Dehm, Biologic and Clinical Significance of Androgen Receptor Variants in Castration Resistant Prostate Cancer, Endocr Relat Cancer, vol.21, issue.4, pp.87-103, 2014.

C. Lu and J. Luo, Decoding the androgen receptor splice variants, Transl Androl Urol, vol.2, issue.3, pp.178-86, 2013.

T. Karantanos, C. Evans, B. Tombal, T. C. Thompson, R. Montironi et al., Understanding the mechanisms of androgen deprivation resistance in prostate cancer at the molecular level, Eur Urol, vol.67, issue.3, pp.470-479, 2015.

J. Luo, Development of AR-V7 as a putative treatment selection marker for metastatic castration-resistant prostate cancer, Asian J Androl, vol.18, issue.4, pp.580-585, 2016.

C. Ciccarese, M. Santoni, M. Brunelli, S. Buti, A. Modena et al., AR-V7 and prostate cancer: The watershed for treatment selection?, Cancer Treat Rev, vol.43, pp.27-35, 2016.

J. Ceraline, E. Erdmann, P. Erbs, M. Deslandres-cruchant, D. Jacqmin et al., A yeast-based functional assay for the detection of the mutant androgen receptor in prostate cancer, Eur J Endocrinol, vol.148, issue.1, pp.99-110, 2003.

F. Cottard, I. Asmane, E. Erdmann, J. Bergerat, J. Kurtz et al., Constitutively Active Androgen Receptor Variants Upregulate Expression of Mesenchymal Markers in Prostate Cancer Cells, PLoS ONE, vol.8, issue.5, p.63466, 2013.

F. Cottard, P. O. Madi-berthélémy, E. Erdmann, F. Schaff-wendling, C. Keime et al., Dual effects of constitutively active androgen receptor and full-length androgen receptor for N-cadherin regulation in prostate cancer, Oncotarget, vol.8, issue.42, pp.72008-72028, 2017.

F. Sun, H. Chen, W. Li, X. Yang, X. Wang et al., Androgen Receptor Splice Variant AR3 Promotes Prostate Cancer via Modulating Expression of Autocrine/Paracrine Factors, J Biol Chem, vol.289, issue.3, pp.1529-1568, 2014.

D. F. Quail and J. A. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nat Med, vol.19, p.1423, 2013.

L. Hui and Y. Chen, Tumor microenvironment: Sanctuary of the devil, Cancer Lett, vol.368, issue.1, pp.7-13, 2015.

Y. Han, Y. Zhang, T. Jia, and Y. Sun, Molecular mechanism underlying the tumor-promoting functions of carcinoma-associated fibroblasts. Tumor Biol, vol.36, pp.1385-94, 2015.

V. S. Lebleu and R. Kalluri, A peek into cancer-associated fibroblasts: origins, functions and translational impact, Dis Model Mech, vol.11, issue.4, p.29447, 2018.

R. Kalluri, The biology and function of fibroblasts in cancer, Nat Rev Cancer, vol.16, p.582, 2016.

D. Öhlund, E. Elyada, and D. Tuveson, Fibroblast heterogeneity in the cancer wound, J Exp Med, vol.211, issue.8, pp.1503-1526, 2014.

K. Shiga, M. Hara, T. Nagasaki, T. Sato, H. Takahashi et al., Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth, Cancers, vol.7, issue.4, pp.2443-58, 2015.

Y. Jung, J. K. Kim, Y. Shiozawa, J. Wang, A. Mishra et al., Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis, Nat Commun, vol.4, p.1795, 2013.

L. Tao, G. Huang, H. Song, Y. Chen, and L. Chen, Cancer associated fibroblasts: An essential role in the tumor microenvironment, Oncol Lett, vol.14, issue.3, pp.2611-2631, 2017.

S. Wen, H. Chang, J. Tian, Z. Shang, Y. Niu et al., Stromal Androgen Receptor Roles in the Development of Normal Prostate, Benign Prostate Hyperplasia, and Prostate Cancer, Am J Pathol, vol.185, issue.2, pp.293-301, 2015.

M. Singh, R. Jha, J. Melamed, E. Shapiro, S. W. Hayward et al., Stromal Androgen Receptor in Prostate Development and Cancer, Am J Pathol, vol.184, issue.10, pp.2598-607, 2014.

P. Cirri and P. Chiarugi, Cancer associated fibroblasts: the dark side of the coin, Am J Cancer Res, vol.1, issue.4, pp.482-97, 2011.

T. Naoko, I. Percy, T. William, K. Nadia, S. Timothy et al., Review of Cancer -Associated Fibroblasts and Therapies that Interfere with Their Activity, Tumor Microenviron Ther, vol.1, p.19, 2013.

H. Shen, X. Yu, F. Yang, Z. Zhang, J. Shen et al., Reprogramming of Normal Fibroblasts into Cancer-Associated Fibroblasts by miRNAs-Mediated CCL2/VEGFA Signaling, PLoS Genet, vol.12, issue.8, p.1006244, 2016.

S. Arena, M. Salati, G. Sorgentoni, F. Barbisan, and M. Orciani, Characterization of tumorderived mesenchymal stem cells potentially differentiating into cancer-associated fibroblasts in lung cancer, Clin Transl Oncol [Internet], 2018.

S. Paget, . The, . Of, and . Growths, CANCER OF THE BREAST. Orig Publ, vol.1, issue.3421, pp.571-574, 1889.

A. F. Olumi, G. D. Grossfeld, S. W. Hayward, P. R. Carroll, T. D. Tlsty et al., Carcinomaassociated Fibroblasts Direct Tumor Progression of Initiated Human Prostatic Epithelium, Cancer Res, vol.59, p.5002, 1999.

G. R. Cunha, P. S. Cooke, and T. Kurita, Role of stromal-epithelial interactions in hormonal responses, Arch Histol Cytol, vol.67, issue.5, pp.417-451, 2004.

W. N. Brennen, S. Chen, S. R. Denmeade, and J. T. Isaacs, Quantification of Mesenchymal Stem Cells (MSCs) at Sites of Human Prostate Cancer, Oncotarget, vol.4, issue.1, pp.106-123, 2013.

D. Öhlund, A. Handly-santana, G. Biffi, E. Elyada, A. S. Almeida et al., Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer, J Exp Med, 2017.

M. R. Junttila and F. J. De-sauvage, Influence of tumour micro-environment heterogeneity on therapeutic response, Nature, vol.501, p.346, 2013.

A. Costa, Y. Kieffer, A. Scholer-dahirel, F. Pelon, B. Bourachot et al., Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer, Cancer Cell, vol.33, issue.3, pp.463-479, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02437793

Y. Sun, X. Mao, C. Fan, C. Liu, A. Guo et al., CXCL12-CXCR4 axis promotes the natural selection of breast cancer cell metastasis, Tumour Biol, vol.35, issue.8, pp.7765-73, 2014.

M. Mckinley, V. O'loughlin, and T. Bidle, Anatomy & Physiology: An Integrative Approach. 2nd Edition. McGraw-Hill Education, 2015.

J. E. Mcneal, Regional morphology and pathology of the prostate, Am J Clin Pathol, vol.49, issue.3, pp.347-57, 1968.

P. Wadhera, An introduction to acinar pressures in BPH and prostate cancer, Nat Rev Urol, vol.10, p.358, 2013.

L. Siegel-rebecca and D. Miller-kimberly, Cancer statistics, vol.68, issue.1, pp.7-30, 2018.

M. Wong, W. B. Goggins, H. Wang, F. Fung, C. Leung et al., Global Incidence and Mortality for Prostate Cancer: Analysis of Temporal Patterns and Trends in 36 Countries, Eur Urol, vol.70, issue.5, pp.862-74, 2016.

P. Grosclaude, A. Belot, D. Marliac, L. Remontet, L. Leone et al., Le cancer de la prostate, évolution de l'incidence et de la mortalité en France entre, Progrès En Urol, vol.25, issue.9, pp.536-578, 1980.

W. J. Catalona, Prostate Cancer Screening, Medical Clinics of North America, vol.102, issue.2, pp.199-214, 2018.

M. Daniyal, Z. A. Siddiqui, M. Akram, H. M. Asif, S. Sultana et al., Epidemiology, etiology, diagnosis and treatment of prostate cancer, Asian Pac J Cancer Prev APJCP, vol.15, issue.22, pp.9575-9583, 2014.

I. J. Powell, F. D. Vigneau, C. H. Bock, J. Ruterbusch, and L. K. Heilbrun, Reducing Prostate Cancer Racial Disparity: Evidence for Aggressive Early Prostate Cancer PSA Testing of African American Men, Cancer Epidemiol Biomarkers Amp Prev, vol.23, issue.8, p.1505, 2014.

M. Terrier, J. E. Terrier, P. Paparel, N. Morel-journel, A. Baldini et al., diagnostic et pronostic du cancer de la prostate. Médecine Nucléaire, vol.41, pp.329-363, 2017.

D. F. Gleason, G. T. Mellinger, L. J. Arduino, J. C. Bailar, L. E. Becker et al., Prediction of Prognosis for Prostatic Adenocarcinoma by Combined Histological Grading and Clinical Staging, J Urol, vol.111, issue.1, pp.58-64, 1974.

J. Gordetsky and J. Epstein, Grading of prostatic adenocarcinoma: current state and prognostic implications, Diagn Pathol, vol.11, p.25, 2016.

P. A. Humphrey, H. Moch, A. L. Cubilla, T. M. Ulbright, and V. E. Reuter, The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part B: Prostate and Bladder Tumours, Eur Urol, vol.70, issue.1, pp.106-125, 2016.

F. Rozet, C. Hennequin, J. Beauval, P. Beuzeboc, L. Cormier et al., Recommandations en onco-urologie 2016-2018 du CCAFU : Cancer de la prostate, Recommandations, vol.27, pp.95-143, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02297771

J. I. Epstein, L. Egevad, M. B. Amin, B. Delahunt, J. R. Srigley et al., International Society of Urological Pathology (ISUP) Consensus Conference, p.125, 2014.

, on Gleason Grading of Prostatic Carcinoma: Definition of Grading Patterns and Proposal for a New Grading System, Am J Surg Pathol, vol.40, issue.2, pp.244-52, 2016.

O. Beahrs, D. Henson, R. Hutter, and B. Kennedy, American Joint Committee on Cancer Staging Manual for Staging of Cancer, J. B. LIPPINCOTT COMPANY Philadelphia, 1992.

B. Mark, K. , C. Peter, L. Mckenney-jesse, K. et al., Prostate cancer -major changes in the American Joint Committee on Cancer eighth edition cancer staging manual, CA Cancer J Clin, vol.67, issue.3, pp.245-53, 2017.

A. V. D'amico, R. Whittington, and S. Malkowicz, Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer, JAMA, vol.280, issue.11, pp.969-74, 1998.

D. Milonas, M. Kin?ius, G. Skul?ius, A. J. Matjo?aitis, I. Gudinavi?ien? et al., Evaluation of D'Amico criteria for low-risk prostate cancer, Scand J Urol, vol.48, issue.4, pp.344-353, 2014.

P. C. Albertsen, D. F. Moore, W. Shih, Y. Lin, H. Li et al., Impact of Comorbidity on Survival Among Men With Localized Prostate Cancer, J Clin Oncol, vol.29, issue.10, pp.1335-1376, 2011.

F. C. Hamdy, J. L. Donovan, J. A. Lane, M. Mason, C. Metcalfe et al., 10-Year Outcomes after Monitoring, Surgery, or Radiotherapy for Localized Prostate Cancer, N Engl J Med, vol.375, issue.15, pp.1415-1439, 2016.

A. Murat, E. Tibet, A. Egemen, and A. Mutlu, Transperitoneal versus extraperitoneal robot-assisted laparoscopic radical prostatectomy: A prospective single surgeon randomized comparative study, Int J Urol, vol.22, issue.10, pp.916-937, 2015.

E. Haglind, S. Carlsson, J. Stranne, A. Wallerstedt, U. Wilderäng et al., Urinary Incontinence and Erectile Dysfunction After Robotic Versus Open Radical Prostatectomy: A Prospective, Controlled, Nonrandomised Trial, Eur Urol, vol.68, issue.2, pp.216-241, 2015.

B. A. Sherer and L. A. Levine, Current management of erectile dysfunction in prostate cancer survivors, Curr Opin Urol, vol.24, issue.4, pp.401-407, 2014.

J. Cosset, J. Hannoun-lévi, D. Peiffert, M. Delannes, P. Pommier et al., Curiethérapie du cancer prostatique par implants permanents : le point en 2013. Curiethérapie Coordonné Par Jean-Michel Hannoun-Lévi, vol.17, pp.111-118, 2013.

W. J. Morris, S. Tyldesley, S. Rodda, R. Halperin, H. Pai et al., Androgen Suppression Combined with Elective Nodal and Dose Escalated Radiation Therapy (the ASCENDE-RT Trial): An Analysis of Survival Endpoints for a Randomized Trial Comparing a Low-Dose-Rate Brachytherapy Boost to a Dose-Escalated External Beam Boost for High-and Intermediate-risk Prostate Cancer, Int J Radiat Oncol, vol.98, issue.2, pp.275-85, 2017.

N. Mottet, J. Bellmunt, M. Bolla, E. Briers, M. G. Cumberbatch et al., EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent, Eur Urol, vol.71, issue.4, pp.618-647, 2017.

W. D. Heemsbergen, A. Al-mamgani, A. Slot, M. Dielwart, and J. V. Lebesque, Longterm results of the Dutch randomized prostate cancer trial: Impact of doseescalation on local, biochemical, clinical failure, and survival, Radiother Oncol, vol.110, issue.1, pp.104-113, 2014.

M. Bolla, P. Maingon, C. C. Villa, S. Kitsios, P. Poortmans et al., Short Androgen Suppression and Radiation Dose Escalation for Intermediate-and High-Risk Localized Prostate Cancer: Results of EORTC Trial 22991, J Clin Oncol, vol.34, issue.15, pp.1748-56, 2016.

D. Dearnaley, I. Syndikus, H. Mossop, V. Khoo, A. Birtle et al., Conventional versus hypofractionated high-dose intensity-modulated radiotherapy for prostate cancer: 5-year outcomes of the randomised, noninferiority, phase 3 CHHiP trial, Lancet Oncol, vol.17, issue.8, pp.1047-60, 2016.

. Huggins-c, J. Stevens-re, and . Hodges-cv, Studies on prostatic cancer: II. The effects of castration on advanced carcinoma of the prostate gland, Arch Surg, vol.43, issue.2, pp.209-232, 1941.

J. W. Moul, Prostate cancer: making the switch from LHRH antagonist to LHRH agonist, Nat Rev Urol, vol.9, issue.3, pp.125-131, 2012.

A. N. Vis, T. M. Van-der-sluis, H. Al-itejawi, R. Van-moorselaar, and E. Meuleman, Risk of disease flare with LHRH agonist therapy in men with prostate cancer: myth or fact? Urol Oncol, vol.33, pp.7-15, 2015.

N. Agarwal, D. Lorenzo, G. Sonpavde, G. Bellmunt, and J. , New agents for prostate cancer, Ann Oncol, vol.25, issue.9, pp.1700-1709, 2014.

P. A. Watson, V. K. Arora, and C. L. Sawyers, Emerging Mechanisms of Resistance to Androgen Receptor Inhibitors in Prostate Cancer, Nat Rev Cancer, vol.15, issue.12, pp.701-712, 2015.

R. Assi, S. Temraz, A. Shamseddine, and D. Mukherji, New Compounds Targeting the Androgen Receptor for Treatment of Advanced Prostate Cancer, Curr Drug Targets, vol.17, issue.3, pp.290-302, 2016.

S. Prekovic, T. Van-den-broeck, S. Linder, M. E. Van-royen, A. Houtsmuller et al., Molecular underpinnings of enzalutamide resistance, Endocr Relat Cancer, 2018.

C. Helsen, T. Van-den-broeck, A. Voet, S. Prekovic, H. Van-poppel et al., Androgen receptor antagonists for prostate cancer therapy, Endocr Relat Cancer, vol.21, issue.4, pp.105-118, 2014.

F. Claessens, C. Helsen, S. Prekovic, T. Van-den-broeck, L. Spans et al., Emerging mechanisms of enzalutamide resistance in prostate cancer, Nat Rev Urol, vol.11, p.712, 2014.

F. Massari, F. Maines, A. Modena, M. Brunelli, E. Bria et al., Castration Resistant Prostate Cancer (CRPC): State of the Art, Perspectives and New Challenges, Former Curr Med Chem -Anti-Cancer Agents, vol.13, issue.6, pp.872-86, 2013.

T. Zhang, J. Zhu, D. J. George, and A. J. Armstrong, Enzalutamide versus abiraterone acetate for the treatment of men with metastatic castration-resistant prostate cancer, Expert Opin Pharmacother, vol.16, issue.4, pp.473-85, 2015.

F. Claessens and L. Moris, The influence of steroid metabolism on CYP17A1 inhibitor activity, Nat Rev Urol, vol.14, p.590, 2017.

A. Thakur, A. Roy, A. Ghosh, M. Chhabra, and S. Banerjee, Abiraterone acetate in the treatment of prostate cancer, Biomed Pharmacother, vol.101, pp.211-219, 2018.

L. C. Harshman and M. Taplin, Abiraterone Acetate: Targeting Persistent Androgen Dependence in Castration-Resistant Prostate Cancer, Adv Ther, vol.30, issue.8, pp.727-774, 2013.

T. Kobayashi, T. Inoue, T. Kamba, and O. Ogawa, Experimental Evidence of Persistent Androgen-Receptor-Dependency in Castration-Resistant Prostate Cancer, Int J Mol Sci, vol.14, issue.8, pp.15615-15650, 2013.

D. J. Mangelsdorf, C. Thummel, M. Beato, P. Herrlich, G. Schutz et al., The nuclear receptor superfamily: the second decade, Cell, vol.83, issue.6, pp.835-844, 1995.

C. Helsen and F. Claessens, Looking at nuclear receptors from a new angle, Mol Cell Endocrinol, vol.382, issue.1, pp.97-106, 2014.

D. Lorente, J. Mateo, Z. Zafeiriou, A. D. Smith, S. Sandhu et al., Switching and withdrawing hormonal agents for castration-resistant prostate cancer, Nat Rev Urol, vol.12, issue.1, pp.37-47, 2015.

I. J. Mcewan, Intrinsic disorder in the androgen receptor: identification, characterisation and drugability, Mol Biosyst, vol.8, issue.1, pp.82-90, 2012.

A. E. Monaghan and I. J. Mcewan, A sting in the tail: the N-terminal domain of the androgen receptor as a drug target, Asian J Androl, vol.18, issue.5, pp.687-94, 2016.

C. Rinaldi, L. C. Bott, and K. H. Fischbeck, Muscle matters in Kennedy's disease. Neuron, vol.82, pp.251-254, 2014.

G. Querin, G. Soraru, and P. Pradat, Kennedy disease (X-linked recessive bulbospinal neuronopathy): A comprehensive review from pathophysiology to therapy, Rev Neurol, vol.173, issue.5, pp.326-363, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01520284

S. Yoo, A. Pettersson, K. M. Jordahl, R. T. Lis, S. Lindstrom et al., Androgen receptor CAG repeat polymorphism and risk of TMPRSS2:ERG-positive prostate 128 cancer, Cancer Epidemiol Biomarkers Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol, vol.23, issue.10, pp.2027-2058, 2014.

X. Zhai, X. Qu, L. Guo, and Q. Ha, Correlation study between the polymorphism of repetitive sequence in gene CAG of androgen receptor and the occurrence and progression of prostate cancer, Asian Pac J Trop Med, vol.7, issue.4, pp.301-305, 2014.

A. Julia, R. Christine, M. Alastair, E. Pascal, N. Yves et al., Reversible Amyloid Fiber Formation in the N Terminus of Androgen Receptor, ChemBioChem, vol.15, issue.16, pp.2370-2373, 2014.

E. Oppong, G. Stier, M. Gaal, R. Seeger, M. Stoeck et al., An Amyloidogenic Sequence at the N-Terminus of the Androgen Receptor Impacts Polyglutamine Aggregation, vol.7, p.44, 2017.

I. J. Mcewan and J. Gustafsson, Interaction of the human androgen receptor transactivation function with the general transcription factor TFIIF, Proc Natl Acad Sci, vol.94, issue.16, p.8485, 1997.

M. E. Tan, J. Li, H. E. Xu, K. Melcher, and Y. E. , Androgen receptor: structure, role in prostate cancer and drug discovery, Acta Pharmacol Sin, vol.36, issue.1, pp.3-23, 2015.

R. A. Davey and M. Grossmann, Androgen Receptor Structure, Function and Biology: From Bench to Bedside, Clin Biochem Rev, vol.37, issue.1, pp.3-15, 2016.

T. Tanner, F. Claessens, and A. Haelens, The hinge region of the androgen receptor plays a role in proteasome-mediated transcriptional activation, Ann N Y Acad Sci, vol.1030, pp.587-92, 2004.

A. Haelens, T. Tanner, S. Denayer, L. Callewaert, and F. Claessens, The hinge region regulates DNA binding, nuclear translocation, and transactivation of the androgen receptor, Cancer Res, vol.67, issue.9, pp.4514-4537, 2007.

L. Clinckemalie, D. Vanderschueren, S. Boonen, and F. Claessens, The hinge region in androgen receptor control, Mol Cell Endocrinol, vol.358, issue.1, pp.1-8, 2012.

M. Nadal, S. Prekovic, N. Gallastegui, C. Helsen, M. Abella et al., Structure of the homodimeric androgen receptor ligand-binding domain, Nat Commun, vol.8, p.14388, 2017.

W. Gao, C. E. Bohl, and J. T. Dalton, Chemistry and Structural Biology of Androgen Receptor, Chem Rev, vol.105, issue.9, pp.3352-70, 2005.

M. E. Van-royen, S. M. Cunha, M. C. Brink, K. A. Mattern, A. L. Nigg et al., Compartmentalization of androgen receptor protein-protein interactions in living cells, J Cell Biol, vol.177, issue.1, pp.63-72, 2007.

F. Claessens, S. Denayer, N. Van-tilborgh, S. Kerkhofs, C. Helsen et al., Diverse roles of androgen receptor (AR) domains in AR-mediated signaling, Nucl Recept Signal, vol.6, p.8, 2008.

S. M. Dehm and D. J. Tindall, Androgen Receptor Structural and Functional Elements: Role and Regulation in Prostate Cancer, Mol Endocrinol, vol.21, issue.12, pp.2855-63, 2007.

J. D. Wilson, The Critical Role of Androgens in Prostate Development. Horm Cancer Breast Prostate, vol.40, pp.577-90, 2011.

O. Hiort, The differential role of androgens in early human sex development, BMC Med, vol.11, pp.152-152, 2013.

L. A. Cooper and S. T. Page, Androgens and prostate disease, Asian J Androl, vol.16, issue.2, pp.248-55, 2014.

N. C. Bennett, R. A. Gardiner, J. D. Hooper, D. W. Johnson, and G. C. Gobe, Molecular cell biology of androgen receptor signalling, Int J Biochem Cell Biol, vol.42, issue.6, pp.813-840, 2010.

P. E. Lonergan and D. J. Tindall, Androgen receptor signaling in prostate cancer development and progression, J Carcinog, vol.10, issue.20, 2011.

E. Pietri, V. Conteduca, D. Andreis, I. Massa, E. Melegari et al., Androgen receptor signaling pathways as a target for breast cancer treatment, Endocr Relat Cancer, vol.23, issue.10, pp.485-498, 2016.

S. Baron, M. Manin, C. Beaudoin, L. Leotoing, Y. Communal et al., Androgen Receptor Mediates Non-genomic Activation of Phosphatidylinositol 3-OH Kinase in Androgen-sensitive Epithelial Cells, J Biol Chem, vol.279, issue.15, pp.14579-86, 2004.

J. C. Zarif and C. K. Miranti, The Importance of Non-Nuclear AR Signaling in Prostate Cancer Progression and Therapeutic Resistance, Cell Signal, vol.28, issue.5, pp.348-56, 2016.

F. Rahman and H. C. Christian, Non-classical actions of testosterone: an update, Trends Endocrinol Metab TEM, vol.18, issue.10, pp.371-379, 2007.

S. R. Hammes and P. J. Davis, Overlapping nongenomic and genomic actions of thyroid hormone and steroids, Best Pract Res Clin Endocrinol Metab, vol.29, issue.4, pp.581-93, 2015.

C. W. Gregory, B. He, R. T. Johnson, O. H. Ford, J. L. Mohler et al., A mechanism for androgen receptor-mediated prostate cancer recurrence after androgen deprivation therapy, Cancer Res, vol.61, issue.11, pp.4315-4324, 2001.

D. Obinata, K. Takayama, S. Takahashi, and S. Inoue, Crosstalk of the Androgen Receptor with Transcriptional Collaborators: Potential Therapeutic Targets for Castration-Resistant Prostate Cancer, vol.9, p.22, 2017.

I. H. Gelman, Androgen Receptor Activation in Castration-Recurrent Prostate Cancer: The Role of Src-Family and Ack1 Tyrosine Kinases, Int J Biol Sci, vol.10, issue.6, pp.620-626, 2014.

Q. Wang, H. Wang, Q. Ju, Z. Ding, X. Ge et al., The co-regulators SRC-1 and SMRT are involved in interleukin-6-induced androgen receptor activation, Eur Cytokine Netw, vol.27, issue.4, pp.108-121, 2016.

V. Perissi and M. G. Rosenfeld, Controlling nuclear receptors: the circular logic of cofactor cycles, Nat Rev Mol Cell Biol, vol.6, issue.7, pp.542-54, 2005.

S. M. Lopez, A. I. Agoulnik, M. Zhang, L. E. Peterson, E. Suarez et al., Nuclear Receptor Corepressor 1 Expression and Output Declines with Prostate Cancer Progression, Clin Cancer Res Off J Am Assoc Cancer Res, vol.22, issue.15, pp.3937-3986, 2016.

M. M. Wong, C. Guo, and J. Zhang, Nuclear receptor corepressor complexes in cancer: mechanism, function and regulation, Am J Clin Exp Urol, vol.2, issue.3, pp.169-87, 2014.

P. Riegman, R. J. Vlietstra, J. Van-der-korput, J. C. Romijn, and J. Trapman, Characterization of the Prostate-specific Antigen gene: A novel human kallikreinlike gene, Biochem Biophys Res Commun, vol.159, issue.1, pp.95-102, 1989.

K. B. Cleutjens, C. C. Van-eekelen, H. A. Van-der-korput, A. O. Brinkmann, and J. Trapman, Two androgen response regions cooperate in steroid hormone regulated activity of the prostate-specific antigen promoter, J Biol Chem, vol.271, issue.11, pp.6379-88, 1996.

C. A. Heinlein and C. Chang, Androgen receptor in prostate cancer, Endocr Rev, vol.25, issue.2, pp.276-308, 2004.

G. M. Yousef and E. P. Diamandis, The New Human Tissue Kallikrein Gene Family: Structure, Function, and Association to Disease, Endocr Rev, vol.22, issue.2, pp.184-204, 2001.

V. Vaisanen, S. Eriksson, K. K. Ivaska, H. Lilja, M. Nurmi et al., Development of sensitive immunoassays for free and total human glandular kallikrein 2, Clin Chem, vol.50, issue.9, pp.1607-1624, 2004.

S. W. Lee, K. Hosokawa, S. Kim, O. C. Jeong, H. Lilja et al., A Highly Sensitive Porous Silicon (P-Si)-Based Human Kallikrein 2 (hK2) Immunoassay Platform toward Accurate Diagnosis of Prostate Cancer. Star A, editor, Sensors, vol.15, issue.5, pp.11972-87, 2015.

B. Lin, C. Ferguson, J. T. White, S. Wang, R. Vessella et al., Prostatelocalized and Androgen-regulated Expression of the Membrane-bound Serine Protease TMPRSS2, Cancer Res, vol.59, issue.17, p.4180, 1999.

S. A. Tomlins, D. R. Rhodes, S. Perner, S. M. Dhanasekaran, R. Mehra et al., Recurrent Fusion of TMPRSS2 and ETS Transcription Factor Genes in Prostate Cancer. Science, vol.310, p.644, 2005.

A. Burdova, J. Bouchal, S. Tavandzis, and Z. Kolar, TMPRSS2-ERG gene fusion in prostate cancer, Biomed Pap, vol.158, issue.4, pp.502-512, 2014.

F. Sanguedolce, A. Cormio, M. Brunelli, D. 'amuri, A. Carrieri et al., Urine TMPRSS2: ERG Fusion Transcript as a Biomarker for Prostate Cancer: Literature Review, Clin Genitourin Cancer, vol.14, issue.2, pp.117-138, 2016.

D. K. Lam, D. Dang, A. N. Flynn, M. Hardt, and B. L. Schmidt, TMPRSS2, a novel membrane-anchored mediator in cancer pain, Pain, vol.156, issue.5, pp.923-953, 2015.

S. Hussein, S. Satturwar, and T. Van-der-kwast, Young-age prostate cancer, J Clin Pathol, vol.68, issue.7, pp.511-516, 2015.

H. Beltran, R. Yelensky, G. M. Frampton, K. Park, S. R. Downing et al., Targeted Next-generation Sequencing of Advanced Prostate Cancer Identifies Potential Therapeutic Targets and Disease Heterogeneity, Eur Urol, vol.63, issue.5, pp.920-926, 2013.

D. Robinson, E. M. Van-allen, Y. Wu, N. Schultz, R. J. Lonigro et al., Integrative Clinical Genomics of Advanced Prostate Cancer, vol.161, pp.1215-1243, 2015.

A. Kumar, I. Coleman, C. Morrissey, X. Zhang, L. D. True et al., Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer, Nat Med, vol.22, p.369, 2016.

S. Carreira, A. Romanel, J. Goodall, E. Grist, R. Ferraldeschi et al., Tumor clone dynamics in lethal prostate cancer, Sci Transl Med, vol.6, issue.254, pp.254-125, 2014.

B. Sahu, M. Laakso, P. Pihlajamaa, K. Ovaska, I. Sinielnikov et al., FoxA1 specifies unique androgen and glucocorticoid receptor binding events in prostate cancer cells, Cancer Res, vol.73, issue.5, pp.1570-80, 2013.

V. K. Arora, E. Schenkein, R. Murali, S. K. Subudhi, J. Wongvipat et al., Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade, Cell, vol.155, issue.6, pp.1309-1331, 2013.

B. Montgomery, H. H. Cheng, J. Drechsler, and E. A. Mostaghel, Glucocorticoids and prostate cancer treatment: friend or foe?, Asian J Androl, vol.16, issue.3, pp.354-362, 2014.

M. P. Edlind and A. C. Hsieh, PI3K-AKT-mTOR signaling in prostate cancer progression and androgen deprivation therapy resistance, Asian J Androl, vol.16, issue.3, pp.378-86, 2014.

B. Bilir, A. O. Osunkoya, . Wiles-wg-4th, S. Sannigrahi, V. Lefebvre et al., SOX4 Is Essential for Prostate Tumorigenesis Initiated by PTEN Ablation, Cancer Res, vol.76, issue.5, pp.1112-1133, 2016.

B. S. Carver, C. Chapinski, J. Wongvipat, H. Hieronymus, Y. Chen et al., Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell, vol.19, pp.575-86, 2011.

C. Foley and N. Mitsiades, Moving Beyond the Androgen Receptor (AR): Targeting AR-Interacting Proteins to Treat Prostate Cancer. Horm Cancer, vol.7, pp.84-103, 2016.

J. Qin, H. Lee, S. Wu, S. Lin, R. B. Lanz et al., Androgen deprivation-induced NCoA2 promotes metastatic and castration-resistant prostate cancer, J Clin Invest, vol.124, issue.11, pp.5013-5039, 2014.

C. Geng, K. Rajapakshe, S. S. Shah, J. Shou, V. K. Eedunuri et al., Androgen Receptor Is the Key Transcriptional Mediator of the Tumor Suppressor SPOP in Prostate Cancer, Cancer Res, vol.74, p.5631, 2014.

N. Lallous, S. V. Volik, S. Awrey, E. Leblanc, R. Tse et al., Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients, Genome Biol, vol.17, issue.1, p.10, 2016.

J. Veldscholte, C. Ris-stalpers, G. G. Kuiper, G. Jenster, C. Berrevoets et al., A mutation in the ligand binding domain of the androgen receptor of human LNCaP cells affects steroid binding characteristics and response to antiandrogens, Biochem Biophys Res Commun, vol.173, issue.2, pp.534-574, 1990.

C. Ris-stalpers, M. C. Verleun-mooijman, J. Trapman, and A. O. Brinkmann, Threonine on amino acid position 868 in the human androgen receptor is essential for androgen binding specificity and functional activity, Biochem Biophys Res Commun, vol.196, issue.1, pp.173-80, 1993.

. Berrevoets-ca, . Umar-a, J. Trapman, and B. Ao, Differential modulation of androgen receptor transcriptional activity by the nuclear receptor co-repressor (N-CoR), Biochem J, vol.379, issue.3, p.731, 2004.

J. D. Joseph, N. Lu, J. Qian, J. Sensintaffar, G. Shao et al., A Clinically Relevant Androgen Receptor Mutation Confers Resistance to Second-Generation Antiandrogens Enzalutamide and ARN-509. Cancer Discov, vol.3, p.1020, 2013.

M. D. Balbas, M. J. Evans, D. J. Hosfield, J. Wongvipat, V. K. Arora et al., Overcoming mutation-based resistance to antiandrogens with rational drug design, Elife, vol.2, p.499, 2009.

A. A. Azad, S. V. Volik, A. W. Wyatt, A. Haegert, L. Bihan et al., Androgen Receptor Gene Aberrations in Circulating Cell-Free DNA: Biomarkers of Therapeutic Resistance in Castration-Resistant Prostate Cancer, Clin Cancer Res, vol.21, issue.10, p.2315, 2015.

S. Prekovic, M. E. Van-royen, A. Voet, B. Geverts, R. Houtman et al., The Effect of F877L and T878A Mutations on Androgen Receptor Response to Enzalutamide, Mol Cancer Ther, vol.15, issue.7, p.1702, 2016.

X. Shi, A. Ma, L. Xia, H. Kung, and R. W. De-vere-white, Functional Analysis of 44 Mutant Androgen Receptors from Human Prostate Cancer, Cancer Res, vol.62, issue.5, p.1496, 2002.

S. N. Maity, M. A. Titus, R. Gyftaki, G. Wu, J. Lu et al., Targeting of CYP17A1 Lyase by VT-464 Inhibits Adrenal and Intratumoral Androgen Biosynthesis and Tumor Growth of Castration Resistant Prostate Cancer, Sci Reports, vol.6, p.35354, 2016.

A. K. Vasudevamurthy, E. Ledet, C. Garvey, B. E. Lewis, and O. Sartor, Estrogen-Mediated Activation of H875Y Androgen Receptor Mutation in a Prostate Cancer Patient, Clin Genitourin Cancer, vol.15, issue.1, pp.111-113, 2017.

X. Y. Zhao, P. J. Malloy, A. V. Krishnan, S. Swami, N. M. Navone et al., Glucocorticoids can promote androgen-independent growth of prostate cancer cells through a mutated androgen receptor, Nat Med, vol.6, issue.6, pp.703-709, 2000.

A. Romanel, G. Tandefelt, D. Conteduca, V. Jayaram, A. Casiraghi et al., Plasma AR and abiraterone-resistant prostate cancer, Sci Transl Med, vol.7, issue.312, pp.312-322, 2015.

A. W. Wyatt, A. A. Azad, S. V. Volik, M. Annala, K. Beja et al., Genomic alterations in cell-free DNA and enzalutamide resistance in castration-resistant prostate cancer, JAMA Oncol, vol.2, issue.12, pp.1598-606, 2016.

T. Hara, J. Miyazaki, H. Araki, M. Yamaoka, N. Kanzaki et al., Novel mutations of androgen receptor: a possible mechanism of bicalutamide withdrawal syndrome, Cancer Res, vol.63, issue.1, pp.149-53, 2003.

H. Liu, A. X. Li, S. Wang, Y. Li, J. Liu et al., Interaction mechanism exploration of Rbicalutamide/S-1 with WT/W741L AR using molecular dynamics simulations, Mol Biosyst, vol.11, issue.12, pp.3347-54, 2015.

H. Liu, R. Han, J. Li, H. Liu, and L. Zheng, Molecular mechanism of R-bicalutamide switching from androgen receptor antagonist to agonist induced by amino acid mutations using molecular dynamics simulations and free energy calculation, J Comput Aided Mol Des, vol.30, issue.12, pp.1189-200, 2016.

C. Guangchun, W. Xiaohui, Z. Shimin, L. Ying, S. Yinghao et al., Androgen receptor mutants detected in recurrent prostate cancer exhibit diverse functional characteristics. The Prostate, vol.63, pp.395-406, 2004.

M. P. Steinkamp, O. A. O'mahony, M. Brogley, H. Rehman, E. W. Lapensee et al., Treatment-Dependent Androgen Receptor Mutations in Prostate Cancer Exploit Multiple Mechanisms to Evade Therapy, Cancer Res, vol.69, issue.10, p.4434, 2009.

S. C. Chan and S. M. Dehm, Constitutive activity of the androgen receptor, Adv Pharmacol San Diego Calif, vol.70, pp.327-66, 2014.

R. Tadokoro-cuccaro, J. Davies, N. P. Mongan, T. Bunch, R. S. Brown et al., Promoter-dependent activity on androgen receptor N-terminal domain mutations in androgen insensitivity syndrome, Sex Dev Genet Mol Biol Evol Endocrinol Embryol Pathol Sex Determ Differ, vol.8, issue.6, pp.339-388, 2014.

J. Bergerat and J. Céraline, Pleiotropic functional properties of androgen receptor mutants in prostate cancer, Hum Mutat, vol.30, issue.2, pp.145-57, 2008.

G. Jenster, J. A. Van-der-korput, J. Trapman, and A. O. Brinkmann, Functional domains of the human androgen receptor, J Steroid Biochem Mol Biol, vol.41, issue.3-8, pp.671-676, 1992.

J. Ceraline, M. D. Cruchant, E. Erdmann, P. Erbs, J. Kurtz et al., Constitutive activation of the androgen receptor by a point mutation in the hinge region: a new mechanism for androgen-independent growth in prostate cancer, Int J Cancer, vol.108, issue.1, pp.152-159, 2004.

S. Cao, Y. Zhan, and Y. Dong, Emerging data on androgen receptor splice variants in prostate cancer, Endocr Relat Cancer, vol.23, issue.12, pp.199-210, 2016.

J. Ceraline, E. Erdmann, P. Erbs, M. Deslandres-cruchant, D. Jacqmin et al., A yeast-based functional assay for the detection of the mutant androgen receptor in prostate cancer, Eur J Endocrinol, vol.148, issue.1, pp.99-110, 2003.

G. Lapouge, G. Marcias, E. Erdmann, P. Kessler, M. Cruchant et al., Specific properties of a C-terminal truncated androgen receptor detected in hormone refractory prostate cancer, Adv Exp Med Biol, vol.617, pp.529-563, 2008.

G. Lapouge, E. Erdmann, G. Marcias, M. Jagla, A. Monge et al., Unexpected paracrine action of prostate cancer cells harboring a new class of androgen receptor mutation--a new paradigm for cooperation among prostate tumor cells, Int J Cancer, vol.121, issue.6, pp.1238-1282, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00189179

X. Ye and R. A. Weinberg, Epithelial-Mesenchymal Plasticity: A central regulator of cancer progression, Trends Cell Biol, vol.25, issue.11, pp.675-86, 2015.

F. Cottard, I. Asmane, E. Erdmann, J. Bergerat, J. Kurtz et al., Constitutively Active Androgen Receptor Variants Upregulate Expression of Mesenchymal Markers in Prostate Cancer Cells, PLoS One, vol.8, issue.5, 2013.

F. Cottard, P. O. Madi-berthélémy, E. Erdmann, F. Schaff-wendling, C. Keime et al., Dual effects of constitutively active androgen receptor and full-length androgen receptor for N-cadherin regulation in prostate cancer, Oncotarget, vol.8, issue.42, pp.72008-72028, 2017.

W. Streicher, F. Zengerling, M. Laschak, W. Weidemann, M. Hopfner et al., AR-Q640X, a model to study the effects of constitutively active C-terminally truncated AR variants in prostate cancer cells, World J Urol, vol.30, issue.3, pp.333-342, 2012.

D. Xu, Y. Zhan, Y. Qi, B. Cao, S. Bai et al., Androgen Receptor Splice Variants Dimerize to Transactivate Target Genes. Cancer Res, vol.75, p.3663, 2015.

W. Streicher, M. Luedeke, A. Azoitei, F. Zengerling, A. Herweg et al., Stilbene Induced Inhibition of Androgen Receptor Dimerization: Implications for AR and AR?LBD-Signalling in Human Prostate Cancer Cells, PLOS ONE, vol.9, issue.6, p.98566, 2014.

L. Yang, C. Lin, J. C. Yang, J. C. Tanasa, B. Li et al., LncRNA-Dependent Mechanisms of Androgen Receptor-regulated Gene Activation Programs, Nature, vol.500, issue.7464, pp.598-602, 2013.

F. Zengerling, A. Azoitei, A. Herweg, F. Jentzmik, and M. V. Cronauer, Inhibition of IGF-1R diminishes transcriptional activity of the androgen receptor and its constitutively active, C-terminally truncated counterparts Q640X and, World J Urol, vol.34, issue.5, pp.633-642, 2016.

R. Da-mota, S. Bailey, S. Strivens, R. A. Hayden, A. L. Douglas et al., LSD1 inhibition attenuates androgen receptor V7 splice variant activation in castration resistant prostate cancer models, Cancer Cell Int, vol.18, issue.1, p.71, 2009.

M. C. Hupe, M. R. Hoda, F. Zengerling, S. Perner, A. S. Merseburger et al., The BET-inhibitor PFI-1 diminishes AR/AR-V7 signaling in prostate cancer cells, World J Urol, 2018.

D. Han, S. Gao, K. Valencia, J. Owiredu, W. Han et al., A novel nonsense mutation in androgen receptor confers resistance to CYP17 inhibitor treatment in prostate cancer, Oncotarget, vol.8, issue.4, pp.6796-808, 2017.

K. Le, B. S. Prabhakar, W. Hong, and L. Li, Alternative splicing as a biomarker and potential target for drug discovery, Acta Pharmacol Sin, vol.36, issue.10, pp.1212-1220, 2015.

S. M. Dehm, L. J. Schmidt, H. V. Heemers, R. L. Vessella, and D. J. Tindall, Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance, Cancer Res, vol.68, issue.13, pp.5469-77, 2008.

Z. Guo, X. Yang, F. Sun, R. Jiang, D. E. Linn et al., A Novel Androgen Receptor Splice Variant Is Upregulated during Prostate Cancer Progression and Promotes Androgen-depletion-resistant Growth, Cancer Res, vol.69, issue.6, pp.2305-2318, 2009.

M. Nakazawa, E. S. Antonarakis, and J. Luo, Androgen Receptor Splice Variants in the Era of Enzalutamide and Abiraterone. Horm Cancer, vol.5, pp.265-73, 2014.

Y. Li, S. C. Chan, L. J. Brand, T. H. Hwang, K. Silverstein et al., Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines, Cancer Res, vol.73, issue.2, pp.483-492, 2013.

Z. Yu, S. Chen, A. G. Sowalsky, O. S. Voznesensky, E. A. Mostaghel et al., Rapid Induction of Androgen Receptor Splice Variants by Androgen Deprivation in Prostate Cancer, Clin Cancer Res Off J Am Assoc Cancer Res, vol.20, issue.6, pp.1590-600, 2014.

E. Hörnberg, E. B. Ylitalo, S. Crnalic, H. Antti, P. Stattin et al., Expression of Androgen Receptor Splice Variants in Prostate Cancer Bone Metastases is Associated with Castration-Resistance and Short Survival, PLoS ONE, vol.6, issue.4, p.19059, 2011.

Y. Qu, B. Dai, D. Ye, Y. Kong, K. Chang et al., Constitutively Active AR-V7 Plays an Essential Role in the Development and Progression of Castration-Resistant Prostate Cancer, Sci Reports, vol.5, p.7654, 2015.

E. Jernberg, E. Thysell, E. Bovinder-ylitalo, S. Rudolfsson, S. Crnalic et al., Characterization of Prostate Cancer Bone Metastases According to Expression Levels of Steroidogenic Enzymes and Androgen Receptor Splice Variants, PLoS ONE, vol.8, issue.11, p.77407, 2013.

S. C. Chan, Y. Li, and S. M. Dehm, Androgen Receptor Splice Variants Activate Androgen Receptor Target Genes and Support Aberrant Prostate Cancer Cell Growth Independent of Canonical Androgen Receptor Nuclear Localization Signal, J Biol Chem, vol.287, issue.23, pp.19736-19785, 2012.

R. Hu, C. Lu, E. A. Mostaghel, S. Yegnasubramanian, M. Gurel et al., Distinct transcriptional programs mediated by the ligand-dependent full-length androgen receptor and its splice variants in castration-resistant prostate cancer, Cancer Res, vol.72, issue.14, pp.3457-62, 2012.

P. A. Watson, Y. F. Chen, M. D. Balbas, J. Wongvipat, N. D. Socci et al., Constitutively active androgen receptor splice variants expressed in castrationresistant prostate cancer require full-length androgen receptor, Proc Natl Acad Sci, vol.107, issue.39, pp.16759-65, 2010.

M. Nakazawa, C. Lu, Y. Chen, C. J. Paller, M. A. Carducci et al., Serial blood-based analysis of AR-V7 in men with advanced prostate cancer, Ann Oncol, vol.26, issue.9, pp.1859-65, 2015.

J. Luo, Development of AR-V7 as a putative treatment selection marker for metastatic castration-resistant prostate cancer, Asian J Androl, vol.18, issue.4, pp.580-585, 2016.

R. J. Andersen, N. R. Mawji, J. Wang, G. Wang, S. Haile et al., Regression of castrate-recurrent prostate cancer by a small-molecule inhibitor of the aminoterminus domain of the androgen receptor, Cancer Cell, vol.17, issue.6, pp.535-581, 2010.

J. Myung, C. A. Banuelos, J. G. Fernandez, N. R. Mawji, J. Wang et al., An androgen receptor N-terminal domain antagonist for treating prostate cancer, J Clin Invest, vol.123, issue.7, pp.2948-60, 2013.

Y. C. Yang, C. A. Banuelos, N. R. Mawji, J. Wang, M. Kato et al., Targeting androgen receptor activation function-1 with EPI to overcome resistance mechanisms in castration-resistant prostate cancer, Clin Cancer Res Off J Am Assoc Cancer Res, vol.22, issue.17, pp.4466-77, 2016.

L. J. Brand, M. E. Olson, P. Ravindranathan, H. Guo, A. M. Kempema et al., EPI-001 is a selective peroxisome proliferator-activated receptor-gamma modulator with inhibitory effects on androgen receptor expression and activity in prostate cancer, Oncotarget, vol.6, issue.6, pp.3811-3835, 2015.

E. De-mol, R. B. Fenwick, C. Phang, V. Buzón, E. Szulc et al., EPI-001, a compound active against castration-resistant prostate cancer, targets transactivation unit 5 of the androgen receptor, ACS Chem Biol, vol.11, issue.9, pp.2499-505, 2016.

F. Sun, H. Chen, W. Li, X. Yang, X. Wang et al., Androgen receptor splice variant AR3 promotes prostate cancer via modulating expression of autocrine/paracrine factors, J Biol Chem, vol.289, issue.3, pp.1529-1568, 2014.

D. F. Quail and J. A. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nat Med, vol.19, p.1423, 2013.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, issue.5, pp.646-74, 2011.

M. R. Junttila and F. J. De-sauvage, Influence of tumour micro-environment heterogeneity on therapeutic response, Nature, vol.501, p.346, 2013.

R. O. Hynes, Extracellular matrix: not just pretty fibrils, Science, vol.326, issue.5957, pp.1216-1225, 2009.

C. Bonnans, J. Chou, and Z. Werb, Remodelling the extracellular matrix in development and disease, Nat Rev Mol Cell Biol, vol.15, issue.12, pp.786-801, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01952416

M. W. Pickup, J. K. Mouw, and V. M. Weaver, The extracellular matrix modulates the hallmarks of cancer, EMBO Rep, vol.15, issue.12, pp.1243-53, 2014.

P. Friedl and S. Alexander, Cancer Invasion and the Microenvironment: Plasticity and Reciprocity, Cell, vol.147, issue.5, pp.992-1009, 2011.

A. Bergamaschi, E. Tagliabue, T. Sorlie, B. Naume, T. Triulzi et al., Extracellular matrix signature identifies breast cancer subgroups with different clinical outcome, J Pathol, vol.214, issue.3, pp.357-67, 2008.

T. Rupp, B. Langlois, M. M. Koczorowska, A. Radwanska, Z. Sun et al., Tenascin-C Orchestrates Glioblastoma Angiogenesis by Modulation of Pro-and Anti-angiogenic Signaling, Cell Reports, vol.17, issue.10, pp.2607-2626, 2016.

G. L. Semenza, Cancer-Stromal Cell Interactions Mediated by Hypoxia-Inducible Factors Promote Angiogenesis, Lymphangiogenesis, and Metastasis. Oncogene, vol.32, pp.4057-63, 2013.

J. Xiong, Q. Yang, J. Li, and S. Zhou, Effects of MDM2 inhibitors on vascular endothelial growth factor-mediated tumor angiogenesis in human breast cancer, Angiogenesis, vol.17, issue.1, pp.37-50, 2014.

Y. Zhao and A. A. Adjei, Targeting Angiogenesis in Cancer Therapy: Moving Beyond Vascular Endothelial Growth Factor. The Oncologist, vol.20, pp.660-73, 2015.

O. Tredan, M. Lacroix-triki, S. Guiu, M. Mouret-reynier, J. Barriere et al., Angiogenesis and tumor microenvironment: bevacizumab in the breast cancer model, Target Oncol, vol.10, issue.2, pp.189-98, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02286714

A. Alitalo and M. Detmar, Interaction of tumor cells and lymphatic vessels in cancer progression, Oncogene, vol.31, p.4499, 2011.

K. E. Hunter, C. Palermo, J. C. Kester, K. Simpson, J. Li et al., Heparanase promotes lymphangiogenesis and tumor invasion in pancreatic neuroendocrine tumors, Oncogene, vol.33, issue.14, pp.1799-808, 2014.

J. A. Joyce and D. T. Fearon, T cell exclusion, immune privilege, and the tumor microenvironment, Science, vol.348, issue.6230, pp.74-80, 2015.

A. Marcus, B. G. Gowen, T. W. Thompson, A. Iannello, M. Ardolino et al., Recognition of tumors by the innate immune system and natural killer cells, Adv Immunol, vol.122, pp.91-128, 2014.

L. Hui and Y. Chen, Tumor microenvironment: Sanctuary of the devil, Cancer Lett, vol.368, issue.1, pp.7-13, 2015.

R. Virchow, Die Cellularpathologie in Ihrer Begründung auf Physiologische und Pathologische Gewebelehre, p.1859

H. F. Dvorak, Tumors: Wounds That Do Not Heal, N Engl J Med, vol.315, issue.26, pp.1650-1659, 1986.

P. Martin and R. Nunan, Cellular and molecular mechanisms of repair in acute and chronic wound healing, Br J Dermatol, vol.173, issue.2, pp.370-378, 2015.

P. Paoli, E. Giannoni, and P. Chiarugi, Anoikis molecular pathways and its role in cancer progression, Biochim Biophys Acta, vol.1833, issue.12, pp.3481-98, 2013.

J. S. Byun and K. Gardner, Wounds That Will Not Heal: Pervasive Cellular Reprogramming in Cancer, Am J Pathol, vol.182, issue.4, pp.1055-64, 2013.

R. Kalluri and M. Zeisberg, Fibroblasts in cancer, Nat Rev Cancer, vol.6, issue.5, pp.392-401, 2006.

G. B. Ryan, W. J. Cliff, G. Gabbiani, C. Irle, P. R. Statkov et al., Myofibroblasts in an avascular fibrous tissue, Lab Investig J Tech Methods Pathol, vol.29, issue.2, pp.197-206, 1973.

G. B. Ryan, W. J. Cliff, G. Gabbiani, C. Irle, D. Montandon et al., Myofibroblasts in human granulation tissue, Hum Pathol, vol.5, issue.1, pp.55-67, 1974.

F. Xing, J. Saidou, and K. Watabe, Cancer associated fibroblasts (CAFs) in tumor microenvironment, Front Biosci Landmark Ed, vol.15, pp.166-79, 2010.

P. Cirri and P. Chiarugi, Cancer associated fibroblasts: the dark side of the coin, Am J Cancer Res, vol.1, issue.4, pp.482-97, 2011.

J. Stanisavljevic, J. Loubat-casanovas, M. Herrera, T. Luque, R. Peña et al., Snail1-Expressing Fibroblasts in the Tumor Microenvironment Display Mechanical Properties That Support Metastasis, Cancer Res, vol.75, issue.2, p.284, 2015.

K. Shiga, M. Hara, T. Nagasaki, T. Sato, H. Takahashi et al., Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth, Cancers, vol.7, issue.4, pp.2443-58, 2015.

T. Nagasaki, M. Hara, H. Nakanishi, H. Takahashi, M. Sato et al., Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction, Br J Cancer, vol.110, issue.2, pp.469-78, 2014.

B. Pan, Q. Liao, Z. Niu, L. Zhou, and Y. Zhao, Cancer-associated fibroblasts in pancreatic adenocarcinoma, Future Oncol Lond Engl, vol.11, issue.18, pp.2603-2613, 2015.

Y. Bai, K. Shang, H. Chen, F. Ding, Z. Wang et al., FGF-1/-3/FGFR4 signaling in cancer-associated fibroblasts promotes tumor progression in colon cancer through Erk and MMP-7, Cancer Sci, vol.106, issue.10, pp.1278-87, 2015.

A. Orimo, P. B. Gupta, D. C. Sgroi, F. Arenzana-seisdedos, T. Delaunay et al., Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion, Cell, vol.121, issue.3, pp.335-383, 2005.

H. Papatheodorou, A. D. Papanastasiou, C. Sirinian, C. Scopa, H. P. Kalofonos et al., Expression patterns of SDF1/CXCR4 in human invasive breast carcinoma and adjacent normal stroma: Correlation with tumor clinicopathological parameters and patient survival, vol.210, pp.662-669, 2014.

M. Herrera, A. Herrera, G. Domínguez, J. Silva, V. García et al., Cancerassociated fibroblast and M2 macrophage markers together predict outcome in colorectal cancer patients, Cancer Sci, vol.104, issue.4, pp.437-481, 2009.

J. Harper and R. Sainson, Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Cancer-Assoc Fibroblasts, vol.25, pp.69-77, 2014.

H. Takahashi, K. Sakakura, R. Kawabata-iwakawa, S. Rokudai, M. Toyoda et al., Immunosuppressive activity of cancer-associated fibroblasts in head and neck squamous cell carcinoma, Cancer Immunol Immunother, vol.64, issue.11, pp.1407-1424, 2015.

N. Erez, S. Glanz, Y. Raz, C. Avivi, and I. Barshack, Cancer Associated Fibroblasts express pro-inflammatory factors in human breast and ovarian tumors, Biochem Biophys Res Commun, vol.437, issue.3, pp.397-402, 2013.

C. Servais and N. Erez, From sentinel cells to inflammatory culprits: cancerassociated fibroblasts in tumour-related inflammation, J Pathol, vol.229, issue.2, pp.198-207, 2013.

S. Dawood, L. Austin, and M. Cristofanilli, Cancer stem cells: implications for cancer therapy, Oncol Williston Park N, vol.28, issue.12, pp.1101-1108, 1110.

F. Lotti, A. M. Jarrar, R. K. Pai, M. Hitomi, J. Lathia et al., Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A, J Exp Med, vol.210, issue.13, pp.2851-72, 2013.

Y. Kinugasa, T. Matsui, and N. Takakura, CD44 Expressed on Cancer-Associated Fibroblasts Is a Functional Molecule Supporting the Stemness and Drug Resistance of Malignant Cancer Cells in the Tumor Microenvironment. STEM CELLS, vol.32, pp.145-56, 2013.

Y. Matsuda, S. Kure, and T. Ishiwata, Nestin and other putative cancer stem cell markers in pancreatic cancer, Med Mol Morphol, vol.45, issue.2, pp.59-65, 2012.

A. Tsuyada, A. Chow, J. Wu, G. Somlo, P. Chu et al., CCL2 mediates crosstalk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells, Cancer Res, vol.72, issue.11, pp.2768-79, 2012.

O. Warburg, On the Origin of Cancer Cells, Science, vol.123, issue.3191, p.309, 1956.

C. D. Gonzalez, S. Alvarez, A. Ropolo, C. Rosenzvit, G. Bagnes et al., Autophagy, Warburg, and Warburg Reverse Effects in Human Cancer, BioMed Res Int, p.926729, 2014.

S. Pavlides, D. Whitaker-menezes, R. Castello-cros, N. Flomenberg, A. K. Witkiewicz et al., The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma, Cell Cycle Georget Tex, vol.8, issue.23, pp.3984-4001, 2009.

E. Paolicchi, F. Gemignani, M. Krstic-demonacos, S. Dedhar, L. Mutti et al., Targeting hypoxic response for cancer therapy, Oncotarget, vol.7, issue.12, pp.13464-78, 2016.

U. E. Martinez-outschoorn, S. Pavlides, D. Whitaker-menezes, K. M. Daumer, J. N. Milliman et al., Tumor cells induce the cancer associated fibroblast phenotype via caveolin-1 degradation: implications for breast cancer and DCIS therapy with autophagy inhibitors, Cell Cycle Georget Tex, vol.9, issue.12, pp.2423-2456, 2010.

C. G. Chen-d, Value of caveolin-1 in cancer progression and prognosis: Emphasis on cancer-associated fibroblasts, human cancer cells and mechanism of caveolin-1 expression (Review), Oncol Lett, vol.8, issue.4, pp.1409-1430, 2014.

C. D. Bovenzi, J. Hamilton, P. Tassone, J. Johnson, D. M. Cognetti et al., Prognostic Indications of Elevated MCT4 and CD147 across Cancer Types: A Meta-Analysis, BioMed Res Int, p.242437, 2015.

Z. Drebert, D. Vlieghere, E. Bridelance, J. , D. Wever et al., Glucocorticoids indirectly decrease colon cancer cell proliferation and invasion via effects on cancer-associated fibroblasts, Exp Cell Res, vol.362, issue.2, pp.332-374, 2018.

J. Tommelein, L. Verset, T. Boterberg, P. Demetter, M. Bracke et al., Cancer-Associated Fibroblasts Connect Metastasis-Promoting Communication in Colorectal Cancer, Front Oncol, vol.5, p.63, 2015.

W. Li, X. Zhang, J. Wang, M. Li, C. Cao et al., TGF?1 in fibroblasts-derived exosomes promotes epithelial-mesenchymal transition of ovarian cancer cells, Oncotarget, vol.8, issue.56, pp.96035-96082, 2017.

B. Jahangiri, M. Khalaj-kondori, E. Asadollahi, and M. Sadeghizadeh, Cancerassociated fibroblasts enhance cell proliferation and metastasis of colorectal cancer SW480 cells by provoking long noncoding RNA UCA1, J Cell Commun Signal [Internet], 2018.

W. Meng, S. Xue, and Y. Chen, The role of CXCL12 in tumor microenvironment, Gene, vol.641, pp.105-115, 2018.

I. J. Fidler and M. L. Kripke, The challenge of targeting metastasis, Cancer Metastasis Rev, vol.34, issue.4, pp.635-676, 2015.

J. Guan, H. Zhang, Z. Wen, Y. Gu, Y. Cheng et al., Retinoic acid inhibits pancreatic cancer cell migration and EMT through the downregulation of IL-6 in cancer associated fibroblast cells, Cancer Lett, vol.345, issue.1, pp.132-141, 2014.

Y. Yu, C. Xiao, L. Tan, Q. Wang, X. Li et al., Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-? signalling, Br J Cancer, vol.110, issue.3, pp.724-756, 2014.

L. Alba-castellón, R. Olivera-salguero, A. Mestre-farrera, R. Peña, M. Herrera et al., Snail1-Dependent Activation of Cancer-Associated Fibroblast Controls Epithelial Tumor Cell Invasion and Metastasis, Cancer Res, vol.76, issue.21, p.6205, 2016.

S. Schmitz, G. Bindea, R. I. Albu, B. Mlecnik, and J. Machiels, Cetuximab promotes epithelial to mesenchymal transition and cancer associated fibroblasts in patients with head and neck cancer, Oncotarget, vol.6, issue.33, pp.34288-99, 2015.

Y. Hassona, N. Cirillo, K. Heesom, E. K. Parkinson, and S. S. Prime, Senescent cancerassociated fibroblasts secrete active MMP-2 that promotes keratinocyte discohesion and invasion, Br J Cancer, vol.111, issue.6, pp.1230-1237, 2009.

C. H. Stuelten, D. Byfield, S. Arany, P. R. Karpova, T. S. Stetler-stevenson et al., Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of, J Cell Sci, vol.118, pp.2143-53, 2005.

A. Taguchi, K. Kawana, K. Tomio, A. Yamashita, Y. Isobe et al., Matrix Metalloproteinase (MMP)-9 in Cancer-Associated Fibroblasts (CAFs) Is Suppressed by Omega-3 Polyunsaturated Fatty Acids In Vitro and In, PLoS ONE, vol.9, issue.2, p.89605, 2014.

E. L. Hastie and D. R. Sherwood, A new front in cell invasion: The invadopodial membrane. Integr Mechano-Chem Signals Invasion, vol.95, pp.441-449, 2016.

S. Goicoechea, R. García-mata, J. Staub, A. Valdivia, L. Sharek et al., Palladin promotes invasion of pancreatic cancer cells by enhancing invadopodia formation in cancer-associated fibroblasts, Oncogene, vol.33, issue.10, pp.1265-73, 2014.

A. Glentis, P. Oertle, P. Mariani, A. Chikina, F. El-marjou et al., Cancerassociated fibroblasts induce metalloprotease-independent cancer cell invasion of the basement membrane, Nat Commun, vol.8, p.924, 2017.

I. Gonzalez-zubeldia, J. Dotor, M. Redrado, A. Bleau, I. Manrique et al., Co-migration of colon cancer cells and CAFs induced by TGF?1 enhances liver metastasis, Cell Tissue Res, vol.359, issue.3, pp.829-868, 2015.

Z. Ao, S. H. Shah, L. M. Machlin, R. Parajuli, P. C. Miller et al., Identification of Cancer-Associated Fibroblasts in Circulating Blood from Patients with Metastatic Breast Cancer, Cancer Res, vol.75, issue.22, p.4681, 2015.

Y. P. Choi, J. H. Lee, M. Gao, B. G. Kim, S. Kang et al., Cancer-associated fibroblast promote transmigration through endothelial brain cells in threedimensional in vitro models, Int J Cancer, vol.135, issue.9, pp.2024-2057, 2014.

J. B. Mccarthy, D. El-ashry, E. A. Turley, and . Hyaluronan, Cancer-Associated Fibroblasts and the Tumor Microenvironment in Malignant Progression, Front Cell Dev Biol, vol.6, p.48, 2018.

D. Öhlund, E. Elyada, and D. Tuveson, Fibroblast heterogeneity in the cancer wound, J Exp Med, vol.211, issue.8, pp.1503-1526, 2014.

M. Berdiel-acer, R. Sanz-pamplona, A. Calon, D. Cuadras, A. Berenguer et al., Differences between CAFs and their paired NCF from adjacent colonic mucosa reveal functional heterogeneity of CAFs, providing prognostic information, Mol Oncol, vol.8, issue.7, pp.1290-305, 2014.

Y. Kojima, A. Acar, E. N. Eaton, K. T. Mellody, C. Scheel et al., Autocrine TGF-? and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts, Proc Natl Acad Sci, vol.107, issue.46, 2010.

M. Cadamuro, G. Nardo, S. Indraccolo, L. Dall'olmo, L. Sambado et al., Platelet-derived growth factor-D and Rho GTPases regulate recruitment of cancer-associated fibroblasts in cholangiocarcinoma, Hepatology, vol.58, issue.3, pp.1042-53, 2013.

K. Shinagawa, Y. Kitadai, M. Tanaka, T. Sumida, M. Onoyama et al., Stroma-directed imatinib therapy impairs the tumor-promoting effect of bone marrow-derived mesenchymal stem cells in an orthotopic transplantation model of colon cancer, Int J Cancer, vol.132, issue.4, pp.813-836, 2013.

L. F. Brown, A. J. Guidi, S. J. Schnitt, L. Van-de-water, M. L. Iruela-arispe et al., Vascular stroma formation in carcinoma in situ, invasive carcinoma, and metastatic carcinoma of the breast, Clin Cancer Res Off J Am Assoc Cancer Res, vol.5, issue.5, pp.1041-56, 1999.

F. Strutz, M. Zeisberg, B. Hemmerlein, B. Sattler, K. Hummel et al., Basic fibroblast growth factor expression is increased in human renal fibrogenesis and may mediate autocrine fibroblast proliferation, Kidney Int, vol.57, issue.4, pp.1521-1559, 2000.

M. Shimoda, S. Principe, H. W. Jackson, V. Luga, H. Fang et al., Loss of the Timp gene family is sufficient for the acquisition of the CAF-like cell state, Nat Cell Biol, vol.16, p.889, 2014.

H. Shen, X. Yu, F. Yang, Z. Zhang, J. Shen et al., Reprogramming of Normal Fibroblasts into Cancer-Associated Fibroblasts by miRNAs-Mediated CCL2/VEGFA Signaling, PLoS Genet, vol.12, issue.8, p.1006244, 2016.

N. Takebe, P. Ivy, W. Timmer, N. Khan, T. Schulz et al., Review of Cancer -Associated Fibroblasts and Therapies that Interfere with Their Activity. Tumor Microenviron Ther, vol.1, pp.19-36, 2013.

P. Cirri and P. Chiarugi, Cancer-associated-fibroblasts and tumour cells: a diabolic liaison driving cancer progression, Cancer Metastasis Rev, vol.31, issue.1-2, pp.195-208, 2012.

S. Madar, I. Goldstein, and V. Rotter, Cancer associated fibroblasts" -more than meets the eye, Trends Mol Med, vol.19, issue.8, pp.447-53, 2013.

P. Gascard and T. D. Tlsty, Carcinoma-associated fibroblasts: orchestrating the composition of malignancy, Genes Dev, vol.30, issue.9, pp.1002-1021, 2016.

M. Elkabets, A. M. Gifford, C. Scheel, B. Nilsson, F. Reinhardt et al., Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice, J Clin Invest, vol.121, issue.2, pp.784-99, 2011.

D. C. Radisky, P. A. Kenny, and M. J. Bissell, Fibrosis and Cancer: Do Myofibroblasts Come Also From Epithelial Cells Via EMT?, J Cell Biochem, vol.101, issue.4, pp.830-839, 2007.

E. M. Zeisberg, S. Potenta, L. Xie, M. Zeisberg, and R. Kalluri, Discovery of Endothelial to Mesenchymal Transition as a Source for Carcinoma-Associated Fibroblasts, Cancer Res, vol.67, issue.21, p.10123, 2007.

A. Nuschke, Activity of mesenchymal stem cells in therapies for chronic skin wound healing, Organogenesis, vol.10, issue.1, pp.29-37, 2014.

W. Zhu, H. Sun, and G. Lyu, Advances in the mechanism of mesenchymal stem cells in promoting wound healing, Chin J Burns, vol.31, issue.6, pp.476-484, 2015.

P. Barcellos-de-souza, V. Gori, F. Bambi, and P. Chiarugi, Tumor microenvironment: bone marrow-mesenchymal stem cells as key players, Biochim Biophys Acta, vol.1836, issue.2, pp.321-356, 2013.

P. J. Mishra, P. J. Mishra, R. Humeniuk, D. J. Medina, G. Alexe et al., Associated Fibroblast Like Differentiation of Human Mesenchymal Stem Cells, Cancer Res, vol.68, issue.11, pp.4331-4340, 2008.

N. C. Direkze, K. Hodivala-dilke, J. R. Hunt, T. Poulsom, R. Oukrif et al., Bone Marrow Contribution to Tumor-Associated Myofibroblasts and Fibroblasts, Cancer Res, vol.64, issue.23, pp.8492-8497, 2004.

M. Quante, S. P. Tu, H. Tomita, T. Gonda, S. S. Wang et al., Bone marrowderived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell, vol.19, pp.257-72, 2011.

Y. Peng, Z. Li, and Z. Li, GRP78 secreted by tumor cells stimulates differentiation of bone marrow mesenchymal stem cells to cancer-associated fibroblasts, Biochem Biophys Res Commun, vol.440, issue.4, pp.558-63, 2013.

Y. Peng, Z. Li, P. Yang, I. P. Newton, H. Ren et al., Direct contacts with colon cancer cells regulate the differentiation of bone marrow mesenchymal stem cells into tumor associated fibroblasts, Biochem Biophys Res Commun, vol.451, issue.1, pp.68-73, 2014.

L. Shangguan, X. Li, Z. Wang, and Z. Luo, Transforming growth factor-beta1 induces bone marrow-derived mesenchymal stem cells to differentiate into cancerassociated fibroblasts, Chin J Oncol, vol.37, issue.11, pp.804-813, 2015.

X. Yang, J. Hao, Y. Mao, J. Cao, R. Zhu et al., bFGF Promotes Migration and Induces Cancer-Associated Fibroblast Differentiation of Mouse Bone Mesenchymal Stem Cells to Promote Tumor Growth, Stem Cells Dev, 2016.

Z. Mi, S. D. Bhattacharya, V. M. Kim, H. Guo, L. J. Talbot et al., Osteopontin promotes CCL5-mesenchymal stromal cell-mediated breast cancer metastasis. Carcinogenesis, vol.32, pp.477-87, 2011.

C. E. Weber, A. N. Kothari, P. Y. Wai, N. Y. Li, J. Driver et al., Osteopontin Mediates an MZF1-TGF-?1-Dependent Transformation of Mesenchymal Stem Cells into Cancer Associated Fibroblasts in Breast Cancer, Oncogene, vol.34, issue.37, pp.4821-4854, 2015.

H. Luo, G. Tu, Z. Liu, and M. Liu, Cancer-associated fibroblasts: A multifaceted driver of breast cancer progression, Cancer Lett, vol.361, issue.2, pp.155-63, 2015.

V. S. Lebleu and R. Kalluri, A peek into cancer-associated fibroblasts: origins, functions and translational impact, Dis Model Mech, vol.11, issue.4, p.29447, 2018.

R. Kalluri, The biology and function of fibroblasts in cancer, Nat Rev Cancer, vol.16, p.582, 2016.

D. Wever, O. , V. Bockstal, M. Mareel, M. Hendrix et al., Carcinomaassociated fibroblasts provide operational flexibility in metastasis. Cancer-Assoc Fibroblasts, vol.25, pp.33-46, 2014.

R. J. Buchsbaum and S. Y. Oh, Breast Cancer-Associated Fibroblasts: Where We Are and Where We Need to Go, Cancers, vol.8, issue.2, p.19, 2016.

M. Wang, J. Zhao, L. Zhang, F. Wei, Y. Lian et al., Role of tumor microenvironment in tumorigenesis, J Cancer, vol.8, issue.5, pp.761-73, 2017.

A. Ostman and M. Augsten, Cancer-associated fibroblasts and tumor growth--bystanders turning into key players, Curr Opin Genet Dev, vol.19, issue.1, pp.67-73, 2009.

M. Augsten, Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment, Front Oncol, vol.4, p.62, 2014.

H. Sugimoto, T. M. Mundel, M. W. Kieran, and R. Kalluri, Identification of fibroblast heterogeneity in the tumor microenvironment, Cancer Biol Ther, vol.5, issue.12, pp.1640-1646, 2006.

Y. Attieh and D. M. Vignjevic, The hallmarks of CAFs in cancer invasion. Integr Mechano-Chem Signals Invasion, vol.95, pp.493-502, 2016.

S. Kuzet and C. Gaggioli, Fibroblast activation in cancer: when seed fertilizes soil, Cell Tissue Res, vol.365, issue.3, pp.607-626, 2016.

D. A. Barron and D. R. Rowley, The reactive stroma microenvironment and prostate cancer progression, Endocr Relat Cancer, vol.19, issue.6, pp.187-204, 2012.

B. Kru?lin, M. Ulamec, and D. Tomas, Prostate cancer stroma: an important factor in cancer growth and progression, Bosn J Basic Med Sci, vol.15, issue.2, pp.1-8, 2015.

Z. Liu, J. Fang, Y. Xiao, Y. Zhao, R. Cui et al., Prognostic role of vascular endothelial growth factor in prostate cancer: a systematic review and meta-analysis, Int J Clin Exp Med, vol.8, issue.2, pp.2289-98, 2015.

E. S. Antonarakis and M. A. Carducci, Targeting angiogenesis for the treatment of prostate cancer, Expert Opin Ther Targets, vol.16, issue.4, pp.365-76, 2012.

N. Ferrara and A. P. Adamis, Ten years of anti-vascular endothelial growth factor therapy, Nat Rev Drug Discov, vol.15, issue.6, pp.385-403, 2016.

M. Hussein, A. , M. Musalam, and A. O. , Phenotypic characterization of the infiltrating immune cells in normal prostate, benign nodular prostatic hyperplasia and prostatic adenocarcinoma, Exp Mol Pathol, 2009.

, Apr, vol.86, issue.2, pp.108-121

B. Gurel, M. S. Lucia, I. M. Thompson, P. J. Goodman, C. M. Tangen et al., Chronic inflammation in benign prostate tissue is associated with high-grade prostate cancer in the placebo arm of the Prostate Cancer Prevention Trial(, Cancer Epidemiol Biomarkers Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol, vol.23, issue.5, pp.847-56, 2014.

C. De-nunzio, G. Kramer, M. Marberger, R. Montironi, W. Nelson et al., The Controversial Relationship Between Benign Prostatic Hyperplasia and Prostate Cancer: The Role of Inflammation, Eur Urol, vol.60, issue.1, pp.106-123, 2011.

S. Davidsson, A. Ohlson, S. Andersson, K. Fall, A. Meisner et al., CD4 helper T cells, CD8 cytotoxic T cells, and FOXP3+ regulatory T cells with respect to lethal prostate cancer, Mod Pathol, vol.26, p.448, 2012.

J. R. Woo, M. A. Liss, M. T. Muldong, K. Palazzi, A. Strasner et al., Tumor infiltrating B-cells are increased in prostate cancer tissue, J Transl Med, vol.12, pp.30-30, 2014.

M. Lanciotti, L. Masieri, M. R. Raspollini, A. Minervini, A. Mari et al., The Role of M1 and M2 Macrophages in Prostate Cancer in relation to Extracapsular Tumor Extension and Biochemical Recurrence after Radical Prostatectomy, BioMed Res Int, p.486798, 2014.

G. Comito, E. Giannoni, C. P. Segura, P. Barcellos-de-souza, M. R. Raspollini et al., Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression, Oncogene, vol.33, issue.19, pp.2423-2454, 2014.

A. F. Olumi, G. D. Grossfeld, S. W. Hayward, P. R. Carroll, T. D. Tlsty et al., Carcinoma-associated Fibroblasts Direct Tumor Progression of Initiated Human Prostatic Epithelium, Cancer Res, vol.59, pp.5002-5013, 1999.

E. Giannoni, F. Bianchini, L. Masieri, S. Serni, T. E. Calorini et al., Reciprocal Activation of Prostate Cancer Cells and Cancer-Associated Fibroblasts Stimulates Epithelial-Mesenchymal Transition and Cancer Stemness, Cancer Res, vol.70, issue.17, p.6945, 2010.

C. Liao, H. Adisetiyo, M. Liang, and P. Roy-burman, Cancer-Associated Fibroblasts Enhance the Gland-Forming Capability of Prostate Cancer Stem Cells, Cancer Res, vol.70, issue.18, pp.7294-303, 2010.

L. A. Geary, K. A. Nash, H. Adisetiyo, M. Liang, C. Liao et al., CAFsecreted Annexin A1 Induces Prostate Cancer Cells to Gain Stem Cell-like Features, Mol Cancer Res MCR, vol.12, issue.4, pp.607-628, 2014.

H. Adisetiyo, M. Liang, C. Liao, J. H. Jeong, M. B. Cohen et al., Dependence of Castration-Resistant Prostate Cancer (CRPC) Stem Cells on CRPC-Associated Fibroblasts, J Cell Physiol, vol.229, issue.9, pp.1170-1176, 2014.

E. Giannoni, F. Bianchini, L. Calorini, and P. Chiarugi, Cancer Associated Fibroblasts Exploit Reactive Oxygen Species Through a Proinflammatory Signature Leading to Epithelial Mesenchymal Transition and Stemness. Antioxidants Redox Signal, vol.14, pp.2361-71, 2011.

M. L. Jones, J. Siddiqui, K. J. Pienta, and R. H. Getzenberg, Circulating fibroblast-like cells in men with metastatic prostate cancer. The Prostate, vol.73, pp.176-81, 2013.

V. Doldi, M. Callari, E. Giannoni, D. 'aiuto, F. Maffezzini et al., Integrated gene and miRNA expression analysis of prostate cancer associated fibroblasts supports a prominent role for interleukin-6 in fibroblast activation, Oncotarget, vol.6, issue.31, pp.31441-60, 2015.

Y. Jung, J. K. Kim, Y. Shiozawa, J. Wang, A. Mishra et al., Recruitment of Mesenchymal Stem Cells Into Prostate Tumors Promotes Metastasis, Nat Commun, vol.4, pp.1795-1795, 2013.

M. A. Castanares, B. T. Copeland, W. H. Chowdhury, M. M. Liu, R. Rodriguez et al., Characterization of a Novel Metastatic Prostate Cancer Cell Line of LNCaP Origin. The Prostate, vol.76, pp.215-240, 2016.

P. Laverman, T. Van-der-geest, S. Terry, D. Gerrits, B. Walgreen et al., Immuno-PET and Immuno-SPECT of Rheumatoid Arthritis with Radiolabeled Anti-Fibroblast Activation Protein Antibody Correlates with Severity of Arthritis, J Nucl Med, vol.56, issue.5, pp.778-83, 2015.

E. J. Hamson, F. M. Keane, S. Tholen, O. Schilling, and M. D. Gorrell, Understanding fibroblast activation protein (FAP): Substrates, activities, expression and targeting for cancer therapy, PROTEOMICS -Clin Appl, vol.8, issue.5-6, pp.454-63, 2014.

R. Dong, J. Guo, Z. Zhang, Y. Zhou, and Y. Hua, Polyphyllin I inhibits gastric cancer cell proliferation by downregulating the expression of fibroblast activation protein alpha (FAP) and hepatocyte growth factor (HGF) in cancer-associated fibroblasts, Biochem Biophys Res Commun, vol.497, issue.4, pp.1129-1163, 2018.

M. L. Wikberg, S. Edin, I. V. Lundberg, B. Van-guelpen, A. M. Dahlin et al., High intratumoral expression of fibroblast activation protein (FAP) in colon cancer is associated with poorer patient prognosis, Tumour Biol, vol.34, issue.2, pp.1013-1033, 2013.

E. Tran, D. Chinnasamy, Z. Yu, R. A. Morgan, C. Lee et al., Immune targeting of fibroblast activation protein triggers recognition of multipotent bone marrow stromal cells and cachexia, J Exp Med, vol.210, issue.6, pp.1125-1160, 2013.

M. F. Garcia, V. Calzada, X. Camacho, E. Goicochea, J. P. Gambini et al., Microwave-assisted Synthesis of HYNIC Protected Analogue for 99mTc Labeled Antibody, Curr Radiopharm, vol.7, issue.2, pp.84-90, 2014.

M. M. Khalil, J. L. Tremoleda, T. B. Bayomy, and W. Gsell, Molecular SPECT Imaging: An Overview, Int J Mol Imaging, p.796025, 2011.

L. Francesconi and . Technetium, Chemistry and Radiopharmaceutical Applications By K. Schwochau (Forschungszentrum, vol.123, pp.12126-12126, 2001.

M. J. Abrams, M. Juweid, C. I. Tenkate, D. A. Schwartz, M. M. Hauser et al., Technetium-99m-Human Polyclonal IgG Radiolabeled via the Hydrazino Nicotinamide Derivative for Imaging Focal Sites of Infection in Rats, J Nucl Med, vol.31, issue.12, pp.2022-2030, 1990.

R. A. Stephenson, C. P. Dinney, K. Gohji, N. G. Ordonez, J. J. Killion et al., Metastatic model for human prostate cancer using orthotopic implantation in nude mice, J Natl Cancer Inst, vol.84, issue.12, pp.951-958, 1992.

Z. El-bitar, R. H. Huesman, R. Buchko, V. Bekaert, D. Brasse et al., A detector response function design in pinhole SPECT including geometrical calibration, Phys Med Biol, vol.58, issue.7, pp.2395-411, 2013.
URL : https://hal.archives-ouvertes.fr/in2p3-00936803

F. Yang, Y. Chen, T. Shen, D. Guo, O. Dakhova et al., Stromal TGF-? signaling induces AR activation in prostate cancer, Oncotarget, vol.5, issue.21, pp.10854-69, 2014.

S. Yu, S. Xia, D. Yang, K. Wang, S. Yeh et al., Androgen receptor in human prostate cancer-associated fibroblasts promotes prostate cancer epithelial cell growth and invasion, Med Oncol, vol.30, issue.3, p.674, 2013.

D. A. Leach, E. F. Need, R. Toivanen, A. P. Trotta, H. M. Palenthorpe et al., Stromal androgen receptor regulates the composition of the microenvironment to influence prostate cancer outcome, Oncotarget, vol.6, issue.18, pp.16135-50, 2015.

P. Tamamis and C. A. Floudas, Elucidating a Key Component of Cancer Metastasis: CXCL12 (SDF-1?) Binding to CXCR4, J Chem Inf Model, vol.54, issue.4, pp.1174-88, 2014.

H. Yamaguchi and R. Sakai, Direct Interaction between Carcinoma Cells and Cancer Associated Fibroblasts for the Regulation of Cancer Invasion, Cancers, vol.7, issue.4, pp.2054-62, 2015.

E. Cortez, P. Roswall, and K. Pietras, Functional subsets of mesenchymal cell types in the tumor microenvironment. Cancer-Assoc Fibroblasts, vol.25, pp.3-9, 2014.

S. M. Ridge, D. Bhattacharyya, E. Dervan, S. D. Naicker, A. J. Burke et al., Secreted factors from metastatic prostate cancer cells stimulate mesenchymal stem cell transition to a pro-tumourigenic "activated" state that enhances prostate cancer cell migration, Int J Cancer, vol.142, issue.10, pp.2056-67, 2018.

A. Costa, Y. Kieffer, A. Scholer-dahirel, F. Pelon, B. Bourachot et al., Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer, Cancer Cell, vol.33, issue.3, pp.463-479, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02437793

V. Paunescu, F. M. Bojin, C. A. Tatu, O. I. Gavriliuc, A. Rosca et al., Tumourassociated fibroblasts and mesenchymal stem cells: more similarities than differences, J Cell Mol Med, vol.15, issue.3, pp.635-681, 2011.

P. D. Johann and I. Müller, Multipotent Mesenchymal Stromal Cells: Possible Culprits in Solid Tumors?, Stem Cells Int, p.914632, 2015.

S. M. Ridge, F. J. Sullivan, and S. A. Glynn, Mesenchymal stem cells: key players in cancer progression. Mol Cancer, vol.16, p.31, 2017.

M. Vizoso, M. Puig, C. Fj, M. Maqueda, A. Velásquez et al., Aberrant DNA methylation in non-small cell lung cancer-associated fibroblasts, Carcinogenesis, vol.36, issue.12, pp.1453-63, 2015.

Q. Xiao, D. Zhou, A. A. Rucki, J. Williams, J. Zhou et al., Cancer associated fibroblasts in pancreatic cancer are reprogrammed by tumor-induced alterations in genomic DNA methylation, Cancer Res, vol.76, issue.18, pp.5395-404, 2016.

J. Yu, K. Walter, N. Omura, S. Hong, Y. A. Li et al., Unlike Pancreatic Cancer Cells Pancreatic Cancer Associated Fibroblasts Display Minimal Gene Induction after 5-Aza-2?-Deoxycytidine, PLoS ONE, vol.7, issue.9, p.43456, 2012.

M. Ruff, A. Leyme, L. Cann, F. Bonnier, D. et al., The Disintegrin and Metalloprotease ADAM12 Is Associated with TGF-?-Induced Epithelial to Mesenchymal Transition, PLoS ONE, vol.10, issue.9, p.139179, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01207281

S. Tyan, C. Hsu, K. Peng, C. Chen, W. Kuo et al., Breast Cancer Cells Induce Stromal Fibroblasts to Secrete ADAMTS1 for Cancer Invasion through an Epigenetic Change, PLoS ONE, vol.7, issue.4, p.35128, 2012.

E. Hedlund and Q. Deng, Single-cell RNA sequencing: Technical advancements and biological applications, vol.59, pp.36-46, 2018.

R. J. Van-soest, J. A. Efstathiou, C. N. Sternberg, and B. Tombal, The Natural History and Outcome Predictors of Metastatic Castration-resistant Prostate Cancer, Eur Urol Focus, vol.2, issue.5, pp.480-487, 2016.

P. J. Tighe, R. R. Ryder, I. Todd, and L. C. Fairclough, ELISA in the multiplex era: Potentials and pitfalls, PROTEOMICS -Clin Appl, vol.9, issue.3-4, pp.406-428, 2015.

S. Riis, A. Stensballe, J. Emmersen, C. P. Pennisi, S. Birkelund et al., Mass spectrometry analysis of adipose-derived stem cells reveals a significant effect of hypoxia on pathways regulating extracellular matrix, Stem Cell Res Ther, vol.7, p.52, 2016.

S. Nath and G. R. Devi, Three-Dimensional Culture Systems in Cancer Research: Focus on Tumor Spheroid Model, Pharmacol Ther, vol.163, pp.94-108, 2016.

T. Eder, A. Weber, H. Neuwirt, G. Grünbacher, C. Ploner et al., Cancer-Associated Fibroblasts Modify the Response of Prostate Cancer Cells to Androgen and Anti-Androgens in Three-Dimensional Spheroid Culture, Int J Mol Sci, vol.17, issue.9, p.1458, 2016.

Y. Zhang, H. Hong, and W. Cai, PET Tracers Based on Zirconium-89, Curr Radiopharm, vol.4, issue.2, pp.131-140, 2011.

L. Spans, C. Helsen, L. Clinckemalie, T. Van-den-broeck, S. Prekovic et al., Comparative Genomic and Transcriptomic Analyses of LNCaP, pp.4-6

, Prostate Cancer Cell Lines. Agoulnik IU, editor. PLoS ONE, vol.9, issue.2, p.90002, 2014.

Y. Du, L. Q. Zhang-l, Y. Shi, . Liu-x, and . Li-x, Curcumin inhibits cancerassociated fibroblast-driven prostate cancer invasion through MAOA/mTOR/HIF-1? signaling, Int J Oncol, vol.47, issue.6, pp.2064-72, 2015.

A. R. Pavan, G. Da-silva, D. H. Jornada, D. E. Chiba, S. Fernandes-gf-dos et al., Unraveling the Anticancer Effect of Curcumin and Resveratrol, Nutrients, vol.8, issue.11, p.628, 2016.

B. C. Jordan, C. D. Mock, R. Thilagavathi, and C. Selvam, Molecular Mechanisms of Curcumin and Its Semisynthetic Analogues in Prostate Cancer Prevention and Treatment, Life Sci, vol.152, pp.135-179, 2016.

Q. Chen, Curcumin-Based Anti-Prostate Cancer Agents, Former Curr Med Chem -Anti-Cancer Agents, vol.15, issue.2, pp.138-56, 2015.

E. Schreyer, P. Barthélémy, and F. Cottard,

F. Berthélémy, J. Schaff-wendling, and . Kurtz, Jocelyn Céraline Med Sci, pp.758-764, 2017.

, Détail ci-après

E. Schreyer, E. Erdmann, A. Maglott-roth, P. Ould-madi, and . Berthélémy,

E. Schreyer, F. Ashtiani, C. Demonacos, L. Mutti, and J. Céraline,

I. Amelio, M. Gostev, R. A. Knight, A. E. Willis, G. Melino et al., DRUGSURV: a resource for repositioning of approved and experimental drugs in oncology based on patient survival information, Cell Death Dis, vol.5, 2014.

E. Bakker, K. Tian, L. Mutti, C. Demonacos, J. M. Schwartz et al., Insight into glucocorticoid receptor signalling through interactome model analysis, PLoS Comput Biol, vol.13, 2017.

M. Gonit, J. Zhang, M. Salazar, H. Cui, A. Shatnawi et al., Hormone depletion-insensitivity of prostate cancer cells is supported by the AR without binding to classical response elements, Mol Endocrinol, vol.25, pp.621-634, 2011.

M. Hussain, B. Stutchbury, K. Tian, R. Atalay, J. Schwartz et al., Applications of p53 interactome analysis to personalised drug discovery, Conference: International Work-Conference on Bioinformatics and Biomedical Engineering, pp.192-203, 2014.

Z. Isik, T. Ersahin, V. Atalay, C. Aykanat, and R. Cetin-atalay, A signal transduction score flow algorithm for cyclic cellular pathway analysis, which combines transcriptome and ChIP-seq data, Mol Biosyst, vol.8, 2012.

S. Klamt, J. Saez-rodriguez, and G. E. , Structural and functional analysis of cellular networks with CellNetAnalyzer, BMC Syst Biol, vol.1, 2007.

T. Mori, M. Flottmann, M. Krantz, T. Akutsu, and K. E. , Stochastic simulation of Boolean rxncon models: towards quantitative analysis of large signaling networks, 2015.

, BMC Syst Biol, vol.9

E. Schreyer, P. Barthélémy, F. Cottard, O. Madi-berthélémy, P. Schaff-wendling et al.,

J. Kurtz and C. J. , Variants du récepteur des androgènes dans le cancer de la prostate, Med Sci, vol.33, pp.758-764, 2017.

P. Shannon, A. Markiel, O. Ozier, N. S. Baliga, J. T. Wang et al., Cytoscape: a software environment for integrated models of biomolecular interaction networks, Genome Res, vol.13, pp.2498-2504, 2003.

D. Szklarczyk, J. H. Morris, H. Cook, M. Kuhn, S. Wyder et al., The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible, Nucleic Acids Res, vol.45, 2017.

M. Tan, J. Li, H. E. Xu, K. Melcher, and Y. , Androgen receptor: structure, role in prostate cancer and drug discovery, Acta Pharmacologica Sinica, vol.36, 2014.

K. Tian, R. Rajendran, M. Doddananjaiah, M. Krstic-demonacos, and J. M. Schwartz, Dynamics of DNA Damage Induced Pathways to Cancer, PLoS One, vol.8, 2013.

H. Werner, G. B. Mills, and R. P. , Cancer Systems Biology: a peak into the future of patient care?, Nat Rev Clin Oncol, vol.11, pp.167-176, 2014.

J. De-l'école-doctorale, , pp.26-27

, EMBO Conference on Nuclear Receptors, pp.24-28

E. Schreyer, E. Erdmann, and F. Cottard,

I. Berthélémy, F. Asmane, J. Wendling, and . Kurtz, Jocelyn Céraline 3) 9ème Forum du Cancéropôle Grand Est, pp.12-13, 2015.

, Poster Androgen receptor variants and microenvironment in prostate cancer

E. Schreyer, E. Erdmann, and F. Cottard,

I. Berthélémy, F. Asmane, J. Wendling, and . Kurtz, Jocelyn Céraline 4) 24ème Journée de l'ARTP, 2015.

, Poster Androgen receptor variants and microenvironment in prostate cancer

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Berthélémy et al.,

, 9th UK Stem Cell Meeting, 2015.

, Poster Androgen receptor variants and microenvironment in prostate cancer

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Berthélémy et al.,

, 4èmes Journées de la FMTS, pp.28-29, 2016.

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Berthélémy et al.,

, 6ème Journée des Récepteurs Nucléaires, vol.9, 2016.

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Berthélémy et al.,

, 23rd Annual Meeting of the European Cancer Center, vol.13, 2016.

, Poster Androgen receptor variants and microenvironment in prostate cancer

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Berthélémy et al.,

. Androgens, , pp.15-17, 2016.

, Poster Androgen receptor variants and microenvironment in prostate cancer

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Berthélémy et al., Jocelyn Céraline 10) 23rd Meeting of the EAU Section of Urological Research, p.20

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, P. Ould-madi et al., Jocelyn Céraline 11) 25ème Journée de l'ARTP, vol.16, 2016.

, Poster Stroma corruption by androgen receptor variants in prostate cancer

E. Schreyer, E. Erdmann, F. Cottard, M. Delbecque, . Ould-madi et al.,

, Forum du Cancéropôle Grand Est, pp.24-25

E. Schreyer, E. Erdmann, . Ould-madi, P. Berthélémy, J. E. Kurtz et al.,

J. De-l'école-doctorale, , pp.22-23

E. Schreyer, E. Erdmann, . Ould-madi, P. Berthélémy, J. E. Kurtz et al., 14) 18ème journée Scientifique Régionale de la Ligue Contre le Cancer, 2017.

, Poster Stroma corruption by androgen receptor variants in prostate cancer

E. Schreyer, E. Erdmann, . Ould-madi, P. Berthélémy, J. E. Kurtz et al., 15) 25rd Meeting of the EAU Section of, Urological Research

E. Schreyer, E. Erdmann, A. Maglott-roth, . Ould-madi, P. Berthélémy et al.,