J. S. De-bono, Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial, Lancet, vol.376, pp.1147-1154, 2010.

J. S. De-bono, Abiraterone and increased survival in metastatic prostate cancer, N. Engl. J. Med, vol.364, 1995.

T. M. Beer, Enzalutamide in metastatic prostate cancer before chemotherapy, N. Engl. J. Med, vol.371, pp.424-433, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02169779

P. W. Kantoff, Sipuleucel-T immunotherapy for castration-resistant prostate cancer, N. Engl. J. Med, vol.363, pp.411-422, 2010.

C. Parker, Alpha emitter radium-223 and survival in metastatic prostate cancer, N. Engl. J. Med, vol.369, pp.213-223, 2013.

D. P. Petrylak, Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer, N. Engl. J. Med, vol.351, pp.1513-1520, 2004.

H. Beltran, Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer, Nat. Med, vol.22, pp.298-305, 2016.

H. Beltran, Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets, Cancer Discov, vol.1, pp.487-495, 2011.

J. I. Epstein, Proposed morphologic classification of prostate cancer with neuroendocrine differentiation, Am. J. Surg. Pathol, vol.38, pp.756-767, 2014.

D. Gao, Organoid cultures derived from patients with advanced prostate cancer, Cell, vol.159, pp.176-187, 2014.

M. B. Lambros, Single-cell analyses of prostate cancer liquid biopsies acquired by apheresis, Clin. Cancer Res, vol.24, pp.5635-5644, 2018.

L. Puca, Patient derived organoids to model rare prostate cancer phenotypes, Nat. Commun, vol.9, p.2404, 2018.

T. Namekawa, K. Ikeda, K. Horie-inoue, and S. Inoue, Application of prostate cancer models for preclinical study: advantages and limitations of cell lines, patient-derived xenografts, and three-dimensional culture of patient-derived cells, Cells, vol.8, p.74, 2019.

L. D. True, A neuroendocrine/small cell prostate carcinoma xenograft-LuCaP 49, Am. J. Pathol, vol.161, pp.705-715, 2002.

J. S. De-bono, Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer, Clin. Cancer Res, vol.14, pp.6302-6309, 2008.

D. Olmos, Circulating tumour cell (CTC) counts as intermediate end points in castration-resistant prostate cancer (CRPC): a single-centre experience, Ann. Oncol, vol.20, pp.27-33, 2009.

C. Massard, Phenotypic and genetic heterogeneity of tumor tissue and circulating tumor cells in patients with metastatic castration-resistant prostate cancer: a report from the PETRUS prospective study, Oncotarget, vol.7, pp.55069-55082, 2016.

G. Attard, Characterization of ERG, AR and PTEN gene status in circulating tumor cells from patients with castration-resistant prostate cancer, Cancer Res, vol.69, pp.2912-2918, 2009.

J. C. Fischer, Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients, Proc. Natl Acad. Sci. USA, vol.110, pp.16580-16585, 2013.

K. C. Andree, Toward a real liquid biopsy in metastatic breast and prostate cancer: diagnostic leukapheresis increases CTC yields in a European prospective multicenter study (CTCTrap), Int. J. Cancer, vol.143, pp.2584-2591, 2018.

T. N. Fehm, Diagnostic leukapheresis for CTC analysis in breast cancer patients: CTC frequency, clinical experiences and recommendations for standardized reporting, Cytometry A, vol.93, pp.1213-1219, 2018.

I. Baccelli, Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay, Nat. Biotechnol, vol.31, pp.539-544, 2013.

C. L. Hodgkinson, Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer, Nat. Med, vol.20, pp.897-903, 2014.

M. R. Girotti, Application of sequencing, liquid biopsies, and patientderived xenografts for personalized medicine in melanoma, Cancer Discov, vol.6, pp.286-299, 2016.

C. J. Morrow, Tumourigenic non-small-cell lung cancer mesenchymal circulating tumour cells: a clinical case study, Ann. Oncol, vol.27, pp.1155-1160, 2016.

L. Cayrefourcq, Establishment and characterization of a cell line from human circulating colon cancer cells, Cancer Res, vol.75, pp.892-901, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01990012

M. Yu, Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility, Science, vol.345, pp.216-220, 2014.

R. Aggarwal, Clinical and genomic characterization of treatmentemergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study, J. Clin. Oncol, vol.36, pp.2492-2503, 2018.

M. Zou, Transdifferentiation as a mechanism of treatment resistance in a mouse model of castration-resistant prostate cancer, Cancer Discov, vol.7, pp.736-749, 2017.

E. Cerami, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data, Cancer Discov, vol.2, pp.401-404, 2012.

J. Gao, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal, Sci. Signal, vol.6, p.1, 2013.

M. E. Legrier, Potentiation of antitumour activity of docetaxel by combination with trastuzumab in a human prostate cancer xenograft model and underlying mechanisms, Br. J. Cancer, vol.96, pp.269-276, 2007.

M. E. Kaighn, K. S. Narayan, Y. Ohnuki, J. F. Lechner, and L. W. Jones, Establishment and characterization of a human prostatic carcinoma cell line (PC-3), Invest. Urol, vol.17, pp.16-23, 1979.

S. Tai, PC3 is a cell line characteristic of prostatic small cell carcinoma, Prostate, vol.71, pp.1668-1679, 2011.

M. Lovf, Multifocal primary prostate cancer exhibits high degree of genomic heterogeneity, Eur. Urol, vol.75, pp.498-505, 2018.

J. G. Lohr, Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer, Nat. Biotechnol, vol.32, pp.479-484, 2014.

S. Y. Ku, Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance, Science, vol.355, pp.78-83, 2017.

A. A. Hamid, Compound genomic alterations of TP53, PTEN, and RB1 tumor suppressors in localized and metastatic prostate cancer, Eur. Urol, vol.77, pp.89-97, 2018.

P. Mu, SOX2 promotes lineage plasticity and antiandrogen resistance in TP53-and RB1-deficient prostate cancer, Science, vol.355, pp.84-88, 2017.

A. Flores-morales, Proteogenomic characterization of patient-derived xenografts highlights the role of REST in neuroendocrine differentiation of castration-resistant prostate cancer, Clin. Cancer Res, vol.25, pp.595-608, 2019.

Y. Gong, U. D. Chippada-venkata, M. D. Galsky, J. Huang, and W. K. Oh, Elevated circulating tissue inhibitor of metalloproteinase 1 (TIMP-1) levels are associated with neuroendocrine differentiation in castration resistant prostate cancer, Prostate, vol.75, pp.616-627, 2015.

N. Xing, J. Qian, D. Bostwick, E. Bergstralh, and C. Y. Young, Neuroendocrine cells in human prostate over-express the anti-apoptosis protein survivin, Prostate, vol.48, pp.7-15, 2001.

C. M. Bielski, Genome doubling shapes the evolution and prognosis of advanced cancers, Nat. Genet, vol.50, pp.1189-1195, 2018.

B. Polzer, Molecular profiling of single circulating tumor cells with diagnostic intention, EMBO Mol. Med, vol.6, pp.1371-1386, 2014.

A. Gnirke, Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing, Nat. Biotechnol, vol.27, pp.182-189, 2009.

M. Martin, Cutadapt removes adapter sequences from high-throughput sequencing reads, EMBnet J, vol.17, p.10, 2011.

G. Khandelwal, Next-generation sequencing analysis and algorithms for PDX and CDX models, Mol. Cancer Res, vol.15, pp.1012-1016, 2017.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics, vol.25, pp.1754-1760, 2009.

A. Tarasov, A. J. Vilella, E. Cuppen, I. J. Nijman, and P. Prins, Sambamba: fast processing of NGS alignment formats, Bioinformatics, vol.31, pp.2032-2034, 2015.

M. A. Depristo, A framework for variation discovery and genotyping using next-generation DNA sequencing data, Nat. Genet, vol.43, pp.491-498, 2011.

G. A. Van-der-auwera, From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline, Curr. Protoc. Bioinformatics, vol.43, p.33, 2013.

K. Cibulskis, Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples, Nat. Biotechnol, vol.31, pp.213-219, 2013.

W. Mclaren, The Ensembl Variant Effect Predictor, Genome Biol, vol.17, p.122, 2016.

D. Tamborero, Cancer Genome Interpreter annotates the biological and clinical relevance of tumor alterations, Genome Med, vol.10, p.25, 2018.

E. S. Venkatraman and A. B. Olshen, A faster circular binary segmentation algorithm for the analysis of array CGH data, Bioinformatics, vol.23, pp.657-663, 2007.

T. Popova, Genome Alteration Print (GAP): a tool to visualize and mine complex cancer genomic profiles obtained by SNP arrays, Genome Biol, vol.10, p.128, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00663915

K. P. Schliep and . Phangorn, phylogenetic analysis in R, vol.27, pp.592-593, 2011.

K. C. Nixon, The Parsimony Ratchet, a new method for rapid parsimony analysis, Cladistics, vol.15, pp.407-414, 1999.

A. Dobin, STAR: ultrafast universal RNA-seq aligner, Bioinformatics, vol.29, pp.15-21, 2013.

S. Anders and W. Huber, Differential expression analysis for sequence count data, Genome Biol, vol.11, p.106, 2010.

L. Wang, S. Wang, and W. Li, RSeQC: quality control of RNA-seq experiments, Bioinformatics, vol.28, pp.2184-2185, 2012.

M. D. Robinson and A. Oshlack, A scaling normalization method for differential expression analysis of RNA-seq data, Genome Biol, vol.11, p.25, 2010.

M. E. Ritchie, limma powers differential expression analyses for RNAsequencing and microarray studies, Nucleic Acids Res, vol.43, p.47, 2015.

C. S. Grasso, The mutational landscape of lethal castration-resistant prostate cancer, Nature, vol.487, pp.239-243, 2012.

S. C. Baca, Punctuated evolution of prostate cancer genomes, Cell, vol.153, pp.666-677, 2013.

D. Robinson, Integrative clinical genomics of advanced prostate cancer, Cell, vol.161, pp.1215-1228, 2015.

A. Kumar, Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer, Nat. Med, vol.22, pp.369-378, 2016.

W. Abida, Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making, JCO Precis. Oncol, 2017.

J. Armenia, The long tail of oncogenic drivers in prostate cancer, Nat. Genet, vol.50, pp.645-651, 2018.

W. Abida, Genomic correlates of clinical outcome in advanced prostate cancer, Proc. Natl Acad. Sci. USA, vol.116, pp.11428-11436, 2019.