T. E. Hutson, B. Escudier, E. E. Bjarnason, G. A. Lim, H. Y. Pittman et al., Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma, J Clin Oncol, vol.32, issue.8, pp.760-767, 2014.

S. Huang, M. A. Bjornsti, and P. J. Houghton, Rapamycins: mechanism of action and cellular resistance, Cancer Biol Ther, vol.2, issue.3, pp.222-232, 2003.

J. M. Barbarino, C. E. Staatz, R. Venkataramanan, and T. E. Klein, Altman RB (2013) PharmGKB summary: cyclosporine and tacrolimus pathways, Pharmacogenet Genomics, vol.23, issue.10, pp.563-585

P. Cai, R. Tsao, and M. E. Ruppen, In vitro metabolic study of temsirolimus: preparation, isolation, and identification of the metabolites, Drug Metab Dispos, vol.35, issue.9, pp.1554-1563, 2007.

A. Lampen, Y. Zhang, I. Hackbarth, L. Z. Benet, and K. F. Sewing, Christians U (1998) Metabolism and transport of the macrolide immunosuppressant sirolimus in the small intestine, J Pharmacol Exp Ther, vol.285, issue.3, pp.1104-1112

, FDA temsirolimus access data, 2017.

K. Klein and U. M. Zanger, Pharmacogenomics of Cytochrome P450 3A4: recent progress toward the "missing heritability, problem. Front Genet, vol.4, p.12, 2013.

D. ?-ochowska, J. Wyzga?, and L. P?czek, Impact of CYP3A4*1B and CYP3A5*3 polymorphisms on the pharmacokinetics of cyclosporine and sirolimus in renal transplant recipients, Ann Transpl, vol.17, issue.3, pp.36-44, 2012.

K. A. Birdwell, B. Decker, J. M. Barbarino, J. F. Peterson, C. M. Stein et al., Clinical pharmacogenetics implementation consortium (CPIC) guidelines for CYP3A5 genotype and tacrolimus dosing, Clin Pharmacol Ther, vol.98, issue.1, pp.19-24, 2015.

C. Marzolini, E. Paus, T. Buclin, and R. B. Kim, Polymorphisms in human MDR1 (P-glycoprotein): recent advances and clinical relevance, Clin Pharmacol Ther, vol.75, issue.1, pp.13-33, 2004.

S. Wolking, E. Schaeffeler, H. Lerche, M. Schwab, and A. T. Nies, Impact of genetic polymorphisms of ABCB1 (MDR1, P-glycoprotein) on drug disposition and potential clinical implications: update of the literature, Clin Pharmacokinet, vol.54, issue.7, pp.709-735, 2015.

Y. Chen, Y. Tang, C. Guo, J. Wang, D. Boral et al., Nuclear receptors in the multidrug resistance through the regulation of drug-metabolizing enzymes and drug transporters, Biochem Pharmacol, vol.83, issue.8, pp.1112-1126, 2012.

A. H. Tolson and H. Wang, Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR, Adv Drug Deliv Rev, vol.62, issue.13, pp.1238-1249, 2010.

G. Bertilsson, J. Heidrich, K. Svensson, M. Asman, L. Jendeberg et al., Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction, Proc Natl Acad Sci, vol.95, issue.21, pp.12208-12213, 1998.

J. Lamba, V. Lamba, and E. Schuetz, Genetic variants of PXR (NR1I2) and CAR (NR1I3) and their implications in drug metabolism and pharmacogenetics, Curr Drug Metab, vol.6, issue.4, pp.369-383, 2005.

K. Benkali, A. Prémaud, N. Picard, J. P. Rérolle, O. Toupance et al., Tacrolimus population pharmacokineticpharmacogenetic analysis and Bayesian estimation in renal transplant recipients, Clin Pharmacokinet, vol.48, issue.12, pp.805-816, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01390737

A. Navarrete, M. P. Martínez-alcázar, I. Durán, E. Calvo, B. Valenzuela et al., Simultaneous online SPE-HPLC-MS/MS analysis of docetaxel, temsirolimus and sirolimus in whole blood and human plasma, J Chromatogr B Analyt Technol Biomed Life Sci, vol.921, pp.35-42, 2013.

J. P. Boni, C. Leister, G. Bender, V. Fitzpatrick, N. Twine et al., Population pharmacokinetics of CCI-779: correlations to safety and pharmacogenomic responses in patients with advanced renal cancer, Clin Pharmacol Ther, vol.77, issue.1, pp.76-89, 2005.

M. Ferraresso, S. Turolo, M. Belinghieri, A. S. Tirelli, P. Grillo et al., The potential of steroids and xenobiotic receptor polymorphisms in forecasting cyclosporine pharmacokinetic variability in young kidney transplant recipients, Pediatr Transpl, vol.16, issue.6, pp.658-663, 2012.

J. B. Woillard, N. Kamar, S. Coste, L. Rostaing, P. Marquet et al., Effect of CYP3A4*22, POR*28, and PPARA rs4253728 on sirolimus in vitro metabolism and trough concentrations in kidney transplant recipients, Clin Chem, vol.59, issue.12, pp.1761-1769, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00852753

L. B. Saltz, J. V. Cox, C. Blanke, L. S. Rosen, L. Fehrenbacher et al., Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer: irinotecan Study Group, N Engl J Med, vol.343, issue.13, pp.905-919, 2000.

M. Ychou, F. Viret, A. Kramar, F. Desseigne, E. Mitry et al., Tritherapy with fluorouracil/leucovorin, irinotecan and oxaliplatin (FOLFIRINOX): a phase II study in colorectal cancer patients with non-resectable liver metastases, Cancer Chemother Pharmacol, vol.62, issue.2, pp.195-201, 2008.

F. Innocenti, R. L. Schilsky, J. Ramírez, L. Janisch, S. Undevia et al., Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer, J Clin Oncol, vol.32, issue.22, pp.2328-2362, 2014.

K. P. Kim, Y. S. Hong, J. L. Lee, K. S. Bae, H. S. Kim et al., A phase I study of UGT1A1 *28/*6 genotype-directed dosing of irinotecan (CPT-11) in Korean patients with metastatic colorectal cancer receiving FOLFIRI, Oncology, vol.88, issue.3, pp.164-72, 2015.

N. F. Smith, W. D. Figg, and A. Sparreboom, Pharmacogenetics of irinotecan metabolism and transport: an update, Toxicol In Vitro, vol.20, issue.2, pp.163-75, 2006.

B. Mohelnikova-duchonova, B. Melichar, and P. Soucek, FOLFOX/ FOLFIRI pharmacogenetics: the call for a personalized approach in colorectal cancer therapy, World J Gastroenterol, vol.20, issue.30, pp.10316-10346, 2014.

D. Lowenberg, C. Whirl-carrillo, M. Ramirez, J. Gong, L. Marsh et al., PharmGKB: irinotecan pathway, 2016.

R. H. Mathijssen, R. J. Van-alphen, J. Verweij, W. J. Loos, K. Nooter et al., Clinical pharmacokinetics and metabolism of irinotecan (CPT-11), Clin Cancer Res, vol.7, issue.8, pp.2182-94, 2001.

E. Beutler, T. Gelbart, and A. Demina, Racial variability in the UDPglucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism?, Proc Natl Acad Sci, vol.95, issue.14, pp.8170-8174, 1998.

Y. Chen, Y. Tang, C. Guo, J. Wang, D. Boral et al., Nuclear receptors in the multidrug resistance through the regulation of drug-metabolizing enzymes and drug transporters, Biochem Pharmacol, vol.83, issue.8, pp.1112-1138, 2012.

A. H. Tolson and H. Wang, Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR, Adv Drug Deliv Rev, vol.62, issue.13, pp.1238-1287, 2010.

J. L. Staudinger, S. Woody, M. Sun, and W. Cui, Nuclear-receptor-mediated regulation of drug-and bile-acid-transporter proteins in gut and liver, Drug Metab Rev, vol.45, issue.1, pp.48-59, 2013.

W. Xie, H. Uppal, S. P. Saini, Y. Mu, J. M. Little et al., Orphan nuclear receptor-mediated xenobiotic regulation in drug metabolism, Drug Discov Today, vol.9, issue.10, pp.442-451, 2004.

B. Blumberg, W. Sabbagh, H. Juguilon, J. Bolado, C. M. Van-meter et al., SXR, a novel steroid and xenobiotic-sensing nuclear receptor, Genes Dev, vol.12, issue.20, pp.3195-205, 1998.

G. Bertilsson, J. Heidrich, K. Svensson, M. Asman, L. Jendeberg et al., Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction, Proc Natl Acad Sci, vol.95, issue.21, pp.12208-12221, 1998.

B. Goodwin, E. Hodgson, D. Costa, D. J. Robertson, G. R. Liddle et al., Transcriptional regulation of the human CYP3A4 gene by the constitutive androstane receptor, Mol Pharmacol, vol.62, issue.2, pp.359-65, 2002.

L. B. Moore, D. J. Parks, S. A. Jones, R. K. Bledsoe, T. G. Consler et al., Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands, J Biol Chem, vol.275, issue.20, pp.15122-15129, 2000.

J. M. Lehmann, D. D. Mckee, M. A. Watson, T. M. Willson, J. T. Moore et al., The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions, J Clin Invest, vol.102, issue.5, pp.1016-1039, 1998.

J. Lamba, V. Lamba, S. Strom, R. Venkataramanan, and E. Schuetz, Novel single nucleotide polymorphisms in the promoter and intron 1 of human pregnane X receptor/NR1I2 and their association with CYP3A4 expression, Drug Metab Dispos, vol.36, issue.1, pp.169-81, 2008.

G. Luo, M. Cunningham, S. Kim, T. Burn, J. Lin et al., CYP3A4 induction by drugs: correlation between a pregnane X receptor reporter gene assay and CYP3A4 expression in human hepatocytes, Drug Metab Dispos, vol.30, issue.7, pp.795-804, 2002.

J. L. Staudinger, C. Xu, Y. J. Cui, and C. D. Klaassen, Nuclear receptormediated regulation of carboxylesterase expression and activity, Expert Opin Drug Metab Toxicol, vol.6, issue.3, pp.261-71, 2010.

N. Albermann, F. H. Schmitz-winnenthal, Z. 'graggen, K. Volk, C. Hoffmann et al., Expression of the drug transporters MDR1/ABCB1, MRP1/ABCC1, MRP2/ABCC2, BCRP/ABCG2, and PXR in peripheral blood mononuclear cells and their relationship with the expression in intestine and liver, Biochem Pharmacol, vol.70, issue.6, pp.949-58, 2005.

A. Evrard and L. Mbatchi, Genetic polymorphisms of drug metabolizing enzymes and transporters: the long way from bench to bedside, Curr Top Med Chem, vol.12, issue.15, pp.1720-1729, 2012.

C. Raynal, J. M. Pascussi, G. Leguelinel, C. Breuker, J. Kantar et al., Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation, Mol Cancer, vol.9, p.46, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00617237

R. H. Mathijssen, J. Verweij, P. De-bruijn, W. J. Loos, A. Sparreboom et al., John's wort on irinotecan metabolism, J Natl Cancer Inst, vol.94, issue.16, pp.1247-1256, 2002.

K. R. Crews, C. F. Stewart, D. Jones-wallace, S. J. Thompson, P. J. Houghton et al., Altered irinotecan pharmacokinetics in pediatric high-grade glioma patients receiving enzyme-inducing anticonvulsant therapy, Clin Cancer Res, vol.8, issue.7, pp.2202-2211, 2002.

J. Lamba, V. Lamba, and E. Schuetz, Genetic variants of PXR (NR1I2) and CAR (NR1I3) and their implications in drug metabolism and pharmacogenetics, Curr Drug Metab, vol.6, issue.4, pp.369-83, 2005.

J. Zhang, P. Kuehl, E. D. Green, J. W. Touchman, P. B. Watkins et al., The human pregnane X receptor: genomic structure and identification and functional characterization of natural allelic variants, Pharmacogenetics, vol.11, issue.7, pp.555-72, 2001.

L. Oleson, L. L. Von-moltke, D. J. Greenblatt, and M. H. Court, Identification of polymorphisms in the 3 0 -untranslated region of the human pregnane X receptor (PXR) gene associated with variability in cytochrome P450 3A (CYP3A) metabolism. Xenobiotica, vol.40, pp.146-62, 2010.

E. Sandanaraj, S. Lal, V. Selvarajan, L. L. Ooi, Z. W. Wong et al., PXR pharmacogenetics: association of haplotypes with hepatic CYP3A4 and ABCB1 messenger RNA expression and doxorubicin clearance in Asian breast cancer patients, NR1I2 and NR1I3 on Pharmacokinetics and Toxicity of Irinotecan 30, vol.14, pp.7116-7142, 2008.

A. Schipani, M. Siccardi, D. 'avolio, A. Baietto, L. Simiele et al., Population pharmacokinetic modeling of the association between 63396C-[T pregnane X receptor polymorphism and unboosted atazanavir clearance, Antimicrob Agents Chemother, vol.54, issue.12, pp.5242-50, 2010.

M. Siccardi, D. 'avolio, A. Baietto, L. Gibbons, S. Sciandra et al., Association of a single-nucleotide polymorphism in the pregnane X receptor (PXR 63396C-[T) with reduced concentrations of unboosted atazanavir, Clin Infect Dis, vol.47, issue.9, pp.1222-1227, 2008.

I. A. Hauser, S. Kruck, S. Gauer, A. T. Nies, S. Winter et al., Human pregnane X receptor genotype of the donor but not of the recipient is a risk factor for delayed graft function after renal transplantation, Clin Pharmacol Ther, vol.91, issue.5, pp.905-921, 2012.

C. F. Xu, N. X. Bing, H. A. Ball, D. Rajagopalan, C. N. Sternberg et al., Pazopanib efficacy in renal cell carcinoma: evidence for predictive genetic markers in angiogenesis-related and exposure-related genes, J Clin Oncol, vol.29, issue.18, pp.2557-64, 2011.

L. C. Mbatchi, A. Schmitt, F. Thomas, Y. Cazaubon, J. Robert et al., Polymorphisms in SLCO1B3 and NR1I2 as genetic determinants of hematotoxicity of carboplatin and paclitaxel combination, Pharmacogenomics, vol.16, issue.13, pp.1439-50, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01913499

M. Swart, H. Whitehorn, Y. Ren, P. Smith, R. S. Ramesar et al., PXR and CAR single nucleotide polymorphisms influence plasma efavirenz levels in South African HIV/AIDS patients, BMC Med Genet, vol.13, p.112, 2012.

A. A. Van-der-veldt, K. Eechoute, H. Gelderblom, J. Gietema, H. J. Guchelaar et al., Genetic polymorphisms associated with a prolonged progression-free survival in patients with metastatic renal cell cancer treated with sunitinib, Clin Cancer Res, vol.17, issue.3, pp.620-629, 2011.

N. P. Van-erp, K. Eechoute, A. A. Van-der-veldt, J. B. Haanen, A. K. Reyners et al., Pharmacogenetic pathway analysis for determination of sunitinib-induced toxicity, J Clin Oncol, vol.27, issue.26, pp.4406-4418, 2009.

S. Poujol, F. Pinguet, M. Ychou, A. G. Abderrahim, J. Duffour et al., A limited sampling strategy to estimate the pharmacokinetic parameters of irinotecan and its active metabolite, SN-38, in patients with metastatic digestive cancer receiving the FOLFIRI regimen, Oncol Rep, vol.18, issue.6, pp.1613-1621, 2007.

S. Poujol, F. Pinguet, F. Malosse, C. Astre, M. Ychou et al., Sensitive HPLC-fluorescence method for irinotecan and four major metabolites in human plasma and saliva: application to pharmacokinetic studies, Clin Chem, vol.49, issue.11, pp.1900-1908, 2003.

E. Gupta, T. M. Lestingi, R. Mick, J. Ramirez, E. E. Vokes et al., Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea, Cancer Res, vol.54, issue.14, pp.3723-3728, 1994.

K. Benkali, A. Prémaud, N. Picard, J. P. Rérolle, O. Toupance et al., Tacrolimus population pharmacokinetic-pharmacogenetic analysis and Bayesian estimation in renal transplant recipients, Clin Pharmacokinet, vol.48, issue.12, pp.805-821, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01390737

M. M. Dring, C. A. Goulding, V. I. Trimble, D. Keegan, A. W. Ryan et al., The pregnane X receptor locus is associated with susceptibility to inflammatory bowel disease, Gastroenterology, vol.130, issue.2, pp.341-349, 2006.

S. Sookoian, G. O. Castaño, A. L. Burgueño, T. F. Gianotti, M. S. Rosselli et al., The nuclear receptor PXR gene variants are associated with liver injury in nonalcoholic fatty liver disease, Pharmacogenet Genom, vol.20, issue.1, pp.1-8, 2010.

T. H. Karlsen, B. A. Lie, K. Frey-frøslie, E. Thorsby, U. Broomé et al., Polymorphisms in the steroid and xenobiotic receptor gene influence survival in primary sclerosing cholangitis, Gastroenterology, vol.131, issue.3, pp.781-788, 2006.

M. Ferraresso, S. Turolo, M. Belinghieri, A. S. Tirelli, P. Grillo et al., The potential of steroids and xenobiotic receptor polymorphisms in forecasting cyclosporine pharmacokinetic variability in young kidney transplant recipients, Pediatr Transpl, vol.16, issue.6, pp.658-63, 2012.

T. Urano, T. Usui, M. Shiraki, Y. Ouchi, and S. Inoue, Association of a single nucleotide polymorphism in the constitutive androstane receptor gene with bone mineral density, Geriatr Gerontol Int, vol.9, issue.3, pp.235-276, 2009.

J. Y. Moon, K. E. Lee, B. C. Chang, E. Jeong, H. Jeong et al., Combined effects of hepatocyte nuclear factor 4a and constitutive androstane receptor on stable warfarin doses, Pharmacogenet Genom, vol.25, issue.1, pp.38-40, 2015.

C. Wyen, H. Hendra, M. Siccardi, M. Platten, H. Jaeger et al., Cytochrome P450 2B6 (CYP2B6) and constitutive androstane receptor (CAR) polymorphisms are associated with early discontinuation of efavirenz-containing regimens, J Antimicrob Chemother, vol.66, issue.9, pp.2092-2100, 2011.

V. Andersen, J. Christensen, A. Ernst, B. A. Jacobsen, A. Tjønneland et al., Polymorphisms in NF-jB, PXR, LXR, PPARc and risk of inflammatory bowel disease, World J Gastroenterol, vol.17, issue.2, pp.197-206, 2011.

D. K. Amre, D. R. Mack, D. Israel, K. Morgan, A. Krupoves et al., Investigation of associations between the pregnane-X receptor gene (NR1I2) and Crohn's disease in Canadian children using a gene-wide haplotype-based approach, Inflamm Bowel Dis, vol.14, issue.9, pp.1214-1222, 2008.

L. Dahan, J. Ciccolini, A. Evrard, L. Mbatchi, J. Tibbitts et al., Sudden death related to toxicity in a patient on capecitabine and irinotecan plus bevacizumab intake: pharmacogenetic implications, J Clin Oncol, vol.30, issue.4, pp.41-45, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01707728

J. R. González, L. Armengol, X. Solé, E. Guinó, J. M. Mercader et al., SNPassoc: an R package to perform whole genome association studies, Bioinformatics, vol.23, issue.5, pp.644-649, 2007.

J. Storey, A. B. Dabney, A. Robinson, and D. , qvalue: Q-value estimation for false discovery rate control, 2016.

B. Yoav and H. Yosef, Controlling the false discovery rate: a practical and powerful approach to multiple testing, J Royal Stat Soc Series B (Methodological), issue.57, pp.289-300, 1995.

J. J. Chen, P. K. Roberson, and M. J. Schell, The false discovery rate: a key concept in large-scale genetic studies, Cancer Control, vol.17, issue.1, pp.58-62, 2010.

J. P. Sinnwell, . Sd, and . Haplo, stats: Statistical analysis of haplotypes with traits and covariates when linkage phase is ambiguous, vol.10, 2015.

. Ncbi and . Dbsnp, Database of single nucleotide polymorphisms (SNPs) and multiple small-scale variations that include insertions/deletions, microsatellites, and non-polymorphic variants, 2015.

S. Marsh and J. M. Hoskins, Irinotecan pharmacogenomics, Pharmacogenomics, vol.11, issue.7, pp.1003-1013, 2010.

R. H. Mathijssen and H. Gurney, Irinogenetics: how many stars are there in the sky?, J Clin Oncol, vol.27, issue.16, pp.2578-2587, 2009.

K. Sai, Y. Saito, N. Tatewaki, M. Hosokawa, N. Kaniwa et al., Association of carboxylesterase 1A genotypes with irinotecan pharmacokinetics in Japanese cancer patients, Br J Clin Pharmacol, vol.70, issue.2, pp.222-255, 2010.

L. E. Carlini, N. J. Meropol, J. Bever, M. L. Andria, T. Hill et al., UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan, Clin Cancer Res, vol.11, issue.3, pp.1226-1262, 2005.

L. C. Mbatchi,

S. Chen, I. Laverdiere, A. Tourancheau, D. Jonker, F. Couture et al., A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients, Pharmacogenomics J, vol.15, issue.6, pp.513-533, 2015.

D. J. Crona, J. Ramirez, W. Qiao, A. J. De-graan, M. J. Ratain et al., Clinical validity of new genetic biomarkers of irinotecan neutropenia: an independent replication study, Pharmacogenomics J, vol.16, issue.1, pp.54-63, 2016.

E. Mc, J. C. Boyer, F. Thomas, S. Quaranta, N. Picard et al., UGT1A1 genotype and irinotecan therapy: general review and implementation in routine practice, Fundam Clin Pharmacol, vol.29, issue.3, pp.219-256, 2015.

J. M. Hoskins, R. M. Goldberg, P. Qu, J. G. Ibrahim, and H. L. Mcleod, UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters, J Natl Cancer Inst, vol.99, issue.17, pp.1290-1295, 2007.

L. Huang, T. Zhang, C. Xie, X. Liao, Q. Yu et al., SLCO1B1 and SLC19A1 gene variants and irinotecan-induced rapid response and survival: a prospective multicenter pharmacogenetics study of metastatic colorectal cancer, PLoS One, vol.8, issue.10, p.77223, 2013.

E. De-mattia, G. Toffoli, J. Polesel, D. 'andrea, M. Corona et al., Pharmacogenetics of ABC and SLC transporters in metastatic colorectal cancer patients receiving first-line FOL-FIRI treatment, Pharmacogenet Genom, vol.23, issue.10, pp.549-57, 2013.

D. A. Kile, S. Mawhinney, C. L. Aquilante, J. E. Rower, J. R. Castillo-mancilla et al., A population pharmacokinetic-pharmacogenetic analysis of atazanavir, AIDS Res Hum Retroviruses, vol.28, issue.10, pp.1227-1261, 2012.

S. Bonora, S. Rusconi, A. Calcagno, M. Bracchi, O. Viganò et al., Successful pharmacogenetics-based optimization of unboosted atazanavir plasma exposure in HIV-positive patients: a randomized, controlled

, J Antimicrob Chemother, vol.70, issue.11, pp.3096-3105, 2015.

, NR1I2 and NR1I3 on Pharmacokinetics and Toxicity of Irinotecan 162

D. B. Longley, W. L. Allen, and P. G. Johnston, « Drug resistance, predictive markers and pharmacogenomics in colorectal cancer, Biochim. Biophys. Acta BBA-Rev. Cancer, vol.1766, issue.2, pp.184-196, 2006.

L. H. Patterson and G. I. Murray, « Tumour cytochrome P450 and drug activation », Curr. Pharm. Des, vol.8, pp.1335-1347, 2002.

N. Widmer, « Review of therapeutic drug monitoring of anticancer drugs part two--targeted therapies », Eur. J. Cancer Oxf. Engl, vol.50, 1990.

T. Chen, Overcoming drug resistance by regulating nuclear receptors, Adv. Drug Deliv. Rev, vol.62, issue.13, pp.1257-1264, 2010.

A. Poso and P. Honkakoski, « Ligand recognition by drug-activated nuclear receptors PXR and CAR: structural, site-directed mutagenesis and molecular modeling studies, Rev. Med. Chem, vol.6, issue.8, pp.937-943, 2006.

Y. Chen, Y. Tang, M. Wang, S. Zeng, and E. D. Nie, « Human pregnane X receptor and resistance to chemotherapy in prostate cancer, Cancer Res, vol.67, pp.10361-10367, 2007.

T. Fujimura, « Clinical significance of steroid and xenobiotic receptor and its targeted gene CYP3A4 in human prostate cancer, Cancer Sci, vol.103, issue.2, p.176, 2012.

B. Zhang, « Pregnane X receptor as a therapeutic target to inhibit androgen activity, Endocrinology, vol.151, pp.5721-5729

I. Heidegger, « Novel therapeutic approaches for the treatment of castration-resistant prostate cancer, J. Steroid Biochem. Mol. Biol, vol.138, pp.248-256, 2013.

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

E. Farber and R. Cameron, « The sequential analysis of cancer development, Adv. Cancer Res, vol.31, pp.125-226, 1980.

S. Puyo, « Recherche d'alternatives thérapeutiques aux taxanes dans les cancers de la prostate de hauts grades: identification d'une signature prédictive de la réponse à l'oxaliplatine, 2011.

I. B. Weinstein, « Carcinogenesis as a multistage process-experimental evidence, IARC Sci. Publ, p.9, 1982.

H. Vainio, P. Magee, D. Mcgregor, and A. J. Mcmichael, Mechanisms of carcinogenesis in risk identification, 1991.

G. I. Evan and K. H. Vousden, « Proliferation, vol.411, pp.342-348, 2001.

I. J. Fidler, D. M. Gersten, and I. R. Hart, The biology of cancer invasion and metastasis, Adv. Cancer Res, vol.28, pp.149-250, 1978.

, Epidémiologie des cancers -Les chiffres du cancer en France | Institut National Du Cancer

D. Sur,

E. D. Crawford, Understanding the epidemiology, natural history, and key pathways involved in prostate cancer, Urology, vol.73, pp.4-10, 2009.

, Evolution de l'incidence et de la mortalité nationales par localisation cancéreuse entre 1980 et, 2012.

W. G. Nelson, A. M. De-marzo, and W. B. Isaacs, N. Engl. J. Med, vol.349, pp.366-381, 2003.

H. Samaratunga, R. A. Gardiner, J. Yaxley, and E. I. Brown, « Atypical prostatic glandular proliferations on needle biopsy: Diagnostic implications, use of immunohistochemistry, and clinical significance, Anal. Quant. Cytol. Histol. Int. Acad. Cytol. Am. Soc. Cytol, vol.28, issue.2, p.104, 2006.

S. Davidsson, « Inflammation, focal atrophic lesions, and prostatic intraepithelial neoplasia with respect to risk of lethal prostate cancer, Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol, vol.20, issue.10, 2011.

J. Felgueiras, J. V. Silva, and M. Fardilha, « Prostate cancer: the need for biomarkers and new therapeutic targets, J. Zhejiang Univ. Sci. B, vol.15, issue.1, p.16, 2014.

S. Bernardini, « Hypermethylation of the CpG islands in the promoter region of the GSTP1 gene in prostate cancer: a useful diagnostic and prognostic marker?, Clin. Chim. Acta Int. J. Clin. Chem, vol.350, issue.2, p.181, 2004.

O. E. Bryzgunova, E. S. Morozkin, S. V. Yarmoschuk, V. V. Vlassov, and P. P. Laktionov, « Methylation-Specific Sequencing of GSTP1 Gene Promoter in Circulating/Extracellular DNA from Blood and Urine of Healthy Donors and Prostate Cancer Patients, Ann. N. Y. Acad. Sci, vol.1137, issue.1, pp.222-225, 2008.

M. Crumbaker, L. Khoja, A. M. Joshua, and «. Ar, Signaling and the PI3K Pathway in Prostate Cancer, Cancers, vol.9, 2017.

B. S. Carver, « Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer, Cancer Cell, vol.19, issue.5, pp.575-586, 2011.

D. J. Mulholland, « Cell Autonomous Role of PTEN in Regulating Castration-Resistant Prostate Cancer Growth, Cancer Cell, vol.19, issue.6, pp.792-804, 2011.

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

B. S. Taylor, « Integrative Genomic Profiling of Human Prostate Cancer, Cancer Cell, vol.18, issue.1, p.11

H. Beltran, « 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.

S. A. Tomlins, « Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer, vol.310, pp.644-648, 2005.

C. E. Barbieri, « The mutational landscape of prostate cancer, Eur. Urol, vol.64, issue.4, pp.567-576, 2013.

M. F. Berger, « The genomic complexity of primary human prostate cancer, Nature, vol.470, 2011.

J. Lindberg, « Exome sequencing of prostate cancer supports the hypothesis of independent tumour origins, Eur. Urol, vol.63, issue.2, pp.347-353, 2013.

R. E. Bakin, D. Gioeli, R. A. Sikes, E. A. Bissonette, and M. J. Weber, « Constitutive activation of the Ras/mitogen-activated protein kinase signaling pathway promotes androgen hypersensitivity in LNCaP prostate cancer cells, Cancer Res, vol.63, issue.8, 2003.

L. Bubendorf, « Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients », Hum. Pathol, vol.31, issue.5, pp.578-583, 2000.

E. S. Martens-uzunova, « Diagnostic and prognostic signatures from the small non-coding RNA transcriptome in prostate cancer, Oncogene, vol.31, issue.8, pp.978-991, 2012.

C. Jerónimo, « Epigenetics in prostate cancer: biologic and clinical relevance, Eur. Urol, vol.60, issue.4, pp.753-766, 2011.

J. W. Catto, « MicroRNA in prostate, bladder, and kidney cancer: a systematic review, Eur. Urol, vol.59, issue.5, pp.671-681, 2011.

K. P. Porkka, M. J. Pfeiffer, K. K. Waltering, R. L. Vessella, T. L. Tammela et al., « MicroRNA expression profiling in prostate cancer, Cancer Res, vol.67, issue.13, pp.6130-6135, 2007.

S. Varambally, « The polycomb group protein EZH2 is involved in progression of prostate cancer, Nature, vol.419, pp.624-629, 2002.

K. Xu, « EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent, Science, vol.338, pp.1465-1469, 2012.

M. Nieto, S. Finn, M. Loda, and W. C. Hahn, Prostate cancer: Re-focusing on androgen receptor signaling, vol.39, pp.1562-1568, 2007.

P. E. Lonergan and D. J. Tindall, et others, « Androgen receptor signaling in prostate cancer development and progression, J. Carcinog, vol.10, issue.1, p.20, 2011.

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

N. Craft, Y. Shostak, M. Carey, and C. L. Sawyers, « A mechanism for hormoneindependent prostate cancer through modulation of androgen receptor signaling by the HER-2/neu tyrosine kinase », Nat. Med, vol.5, issue.3, pp.280-285, 1999.

G. Buchanan, N. M. Greenberg, H. I. Scher, J. M. Harris, V. R. Marshall et al., « Collocation of androgen receptor gene mutations in prostate cancer, Clin. Cancer Res, vol.7, issue.5, pp.1273-1281, 2001.

B. J. Feldman and D. Feldman, « The development of androgen-independent prostate cancer, Nat. Rev. Cancer, vol.1, issue.1, pp.34-45, 2001.

H. Kang, H. Lin, Y. Hu, S. Yeh, K. Huang et al., « From transforming growth factor-? signaling to androgen action: identification of Smad3 as an androgen receptor coregulator in prostate cancer cells, Proc. Natl. Acad. Sci, vol.98, pp.3018-3023, 2001.

H. Meyer, I. Ahrens-fath, A. Sommer, and B. Haendler, « Novel molecular aspects of prostate carcinogenesis, Biomed. Pharmacother, vol.58, issue.1, pp.10-16, 2004.

«. Urofrance, Résultats de la recherche

F. H. Schröder, C. Gosselaar, S. Roemeling, R. Postma, M. J. Roobol et al., PSA and the detection of prostate cancer after 2005. Part I », vol.4, pp.2-12, 2006.

N. Lumen, « Screening and early diagnosis of prostate cancer: an update, Acta Clin. Belg, vol.67, issue.4, pp.270-275, 2012.

D. F. Gleason, « Histologic grading of prostate cancer: a perspective, Hum. Pathol, vol.23, issue.3, pp.273-279, 1992.

L. Egevad, B. Delahunt, J. R. Srigley, and H. Samaratunga, « International Society of Urological Pathology (ISUP) grading of prostate cancer -An ISUP consensus on contemporary grading, APMIS Acta Pathol. Microbiol. Immunol. Scand, vol.124, issue.6, pp.433-435, 2016.

N. Mottet, « EAU Guidelines on Prostate Cancer. Part II: Treatment of Advanced, Relapsing, and Castration-Resistant Prostate Cancer, Eur. Urol, vol.59, pp.572-583, 2011.

A. H. Reid, G. Attard, E. Barrie, J. S. De, and . Bono, « CYP17 inhibition as a hormonal strategy for prostate cancer, Nat. Clin. Pract. Urol, vol.5, issue.11, pp.610-620, 2008.

C. J. Ryan, Abiraterone in Metastatic Prostate Cancer without Previous Chemotherapy, vol.368, pp.138-148, 2013.

C. Tran, « Development of a second-generation antiandrogen for treatment of advanced prostate cancer, vol.324, pp.787-790, 2009.

H. I. Scher, « Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1-2 study, The Lancet, vol.375, pp.1437-1446, 2010.

H. I. Scher, « Increased Survival with Enzalutamide in Prostate Cancer after Chemotherapy, N. Engl. J. Med, vol.367, pp.1187-1197, 2012.

A. Heidenreich, « Cabazitaxel plus prednisone for metastatic castration-resistant prostate cancer progressing after docetaxel: results from the German compassionate-use programme, Eur. Urol, vol.63, issue.6, pp.977-982, 2013.

K. Fizazi, « Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study, The Lancet, vol.377, pp.813-822, 2011.

E. J. Small, « Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer, J. Clin. Oncol, vol.24, pp.3089-3094, 2006.

P. W. Kantoff, « Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer, J. Clin. Oncol, vol.28, issue.7, pp.1099-1105, 2010.

N. Finnström, « Detection of cytochrome P450 mRNA transcripts in prostate samples by RT-PCR », Eur. J. Clin. Invest, vol.31, issue.10, p.880, 2001.

L. Mbatchi, Pharmacogénétique des xénorécepteurs PXR et CAR: implication dans la toxicité hématologique de la chimiothérapie à base de carboplatine et paclitaxel et dans la pharmacocinétique de l'irinotécan. », 2013.

H. Kusuhara and Y. Sugiyama, « In vitro-in vivo extrapolation of transporter-mediated clearance in the liver and kidney, Drug Metab. Pharmacokinet, vol.24, issue.1, pp.37-52, 2009.

U. Gether, P. H. Andersen, O. M. Larsson, and A. Schousboe, « Neurotransmitter transporters: molecular function of important drug targets, Trends Pharmacol. Sci, vol.27, issue.7, p.375, 2006.

I. Spriet, W. Meersseman, J. De-hoon, S. Von-winckelmann, A. Wilmer et al., « Mini-series: II. clinical aspects. clinically relevant CYP450-mediated drug interactions in the ICU », Intensive Care Med, vol.35, issue.4, p.603, 2009.

M. Michael and M. M. Doherty, « Tumoral Drug Metabolism: Overview and Its Implications for Cancer Therapy, J. Clin. Oncol, vol.23, issue.1, pp.205-229, 2005.

J. K. Lamba, Y. S. Lin, E. G. Schuetz, and K. E. Thummel, « Genetic contribution to variable human CYP3A-mediated metabolism, Adv. Drug Deliv. Rev, vol.54, issue.10, p.1271, 2002.

N. Plant, The human cytochrome P450 sub-family: transcriptional regulation, vol.1770, p.478, 2007.

N. E. Sládek, « Aldehyde dehydrogenase-mediated cellular relative insensitivity to the oxazaphosphorines », Curr. Pharm. Des, vol.5, issue.8, pp.607-625, 1999.

J. K. Lamba, Genetic factors influencing cytarabine therapy, vol.10, pp.1657-1674, 2009.

Z. Lu, R. Zhang, and R. B. Diasio, « Purification and characterization of dihydropyrimidine dehydrogenase from human liver. », J. Biol. Chem, vol.267, pp.17102-17109, 1992.

T. K. Kiang, M. H. Ensom, and T. K. Chang, UDP-glucuronosyltransferases and clinical drug-drug interactions, vol.106, pp.97-132, 2005.

P. I. Mackenzie, « Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily », Pharmacogenet. Genomics, vol.15, issue.10, p.677, 2005.

R. G. Deeley, C. Westlake, and S. P. Cole, « Transmembrane transport of endo-and xenobiotics by mammalian ATP-binding cassette multidrug resistance proteins, Physiol. Rev, vol.86, issue.3, p.849, 2006.

A. A. Stavrovskaya and T. P. Stromskaya, « Transport proteins of the ABC family and multidrug resistance of tumor cells, Biochem. Biokhimiia, vol.73, issue.5, pp.592-604, 2008.

Y. G. Assaraf and M. J. Borgnia, « Probing the interaction of the multidrug-resistance phenotype with the polypeptide ionophore gramicidin D via functional channel formation », Eur. J. Biochem, vol.222, issue.3, pp.813-824, 1994.

M. J. Borgnia, G. D. Eytan, and Y. G. Assaraf, « Competition of hydrophobic peptides, cytotoxic drugs, and chemosensitizers on a common P-glycoprotein pharmacophore as revealed by its ATPase activity, J. Biol. Chem, vol.271, issue.6, pp.3163-3171, 1996.

L. Ma, « Design and synthesis of novel 1, 2, 3-triazole-pyrimidine-urea hybrids as potential anticancer agents, Bioorg. Med. Chem. Lett, vol.25, issue.5, pp.1124-1128, 2015.

R. Didziapetris, P. Japertas, A. Avdeef, and A. Petrauskas, « Classification analysis of P-glycoprotein substrate specificity, J. Drug Target, vol.11, issue.7, pp.391-406, 2003.

C. F. Higgins and M. M. Gottesman, « Is the multidrug transporter a flippase?, Trends Biochem. Sci, vol.17, issue.1, pp.18-21, 1992.

M. Dong, F. Penin, and L. G. Baggetto, « Efficient purification and reconstitution of Pglycoprotein for functional and structural studies, J. Biol. Chem, vol.271, pp.28875-28883, 1996.

H. N. Tinsley, « Colon Tumor Cell Growth-Inhibitory Activity of Sulindac Sulfide and Other Nonsteroidal Anti-Inflammatory Drugs Is Associated with Phosphodiesterase 5 Inhibition, Cancer Prev. Res. (Phila. Pa.), vol.3, issue.10, pp.1303-1313, 2010.

L. A. Doyle and D. D. Ross, Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2) », Oncogene, vol.22, pp.7340-7358, 2003.

M. Munoz, M. Henderson, M. Haber, and M. Norris, Role of the MRP1/ABCC1 multidrug transporter protein in cancer, vol.59, pp.752-757, 2007.

S. A. Wardlaw, K. J. Nikula, D. A. Kracko, G. L. Finch, J. R. Thornton-manning et al., Effect of cigarette smoke on CYP1A1, CYP1A2 and CYP2B1/2 of nasal mucosae in F344 rats, Carcinogenesis, vol.19, issue.4, p.655, 1998.

T. K. Chang, « Activation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) by herbal medicines », AAPS J, vol.11, issue.3, p.590, 2009.

N. Isoherranen and K. E. Thummel, « Drug Metabolism and Transport During Pregnancy: How Does Drug Disposition Change during Pregnancy and What Are the Mechanisms that Cause Such Changes?, Drug Metab. Dispos, vol.41, issue.2, p.256, 2013.

J. Gillet and M. M. Gottesman, « Mechanisms of Multidrug Resistance in Cancer, Multi-Drug Resistance in Cancer, pp.47-76, 2010.

S. Rendic and F. J. Carlo, « Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors, vol.29, pp.413-580, 1997.

A. A. Desai, F. Innocenti, and M. J. Ratain, « UGT pharmacogenomics: implications for cancer risk and cancer therapeutics, Pharmacogenet. Genomics, vol.13, issue.8, pp.517-523, 2003.

F. P. Guengerich, Roles of cytochrome P-450 enzymes in chemical carcinogenesis and cancer chemotherapy, Cancer Res, vol.48, issue.11, pp.2946-2954, 1988.

T. Suzuki, K. Nishio, and E. S. Tanabe, « The MRP family and anticancer drug metabolism, Curr. Drug Metab, vol.2, p.367, 2001.

G. I. Murray, « The role of cytochrome P450 in tumour development and progression and its potential in therapy, J. Pathol, vol.192, issue.4, pp.419-426, 2000.

G. I. Murray, M. C. Taylor, M. D. Burke, and W. T. Melvin, « Enhanced expression of cytochrome P450 in stomach cancer. », Br. J. Cancer, vol.77, issue.7, p.1040, 1998.

L. Massaad, « Comparison of mouse and human colon tumors with regard to phase I and phase II drug-metabolizing enzyme systems, Cancer Res, vol.52, issue.23, pp.6567-6575, 1992.

S. V. Bhagwat, M. R. Boyd, and V. Ravindranath, « Multiple forms of cytochrome P450 and associated monooxygenase activities in human brain mitochondria, Biochem. Pharmacol, vol.59, issue.5, pp.573-582, 2000.

H. K. Crewe, L. M. Notley, R. M. Wunsch, M. S. Lennard, and E. M. Gillam, Metabolism of Tamoxifen by Recombinant Human Cytochrome P450 Enzymes: Formation of the 4-Hydroxy, 4?-Hydroxy andN-Desmethyl Metabolites and Isomerization oftrans-4-Hydroxytamoxifen, vol.30, pp.869-874, 2002.

F. J. Gonzalez and H. V. Gelboin, Role of human cytochromes P450 in the metabolic activation of chemical carcinogens and toxins, Drug Metab. Rev, vol.26, issue.2, pp.165-183, 1994.

H. Xie, S. Lundgren, U. Broberg, N. Finnström, A. Rane et al., « Effect of cyclophosphamide on gene expression of cytochromes P450 and ?-actin in the HL-60 cell line », Eur. J. Pharmacol, vol.449, issue.3, pp.197-205, 2002.

M. C. Mcfadyen, H. L. Mcleod, F. C. Jackson, W. T. Melvin, J. Doehmer et al., Cytochrome P450 CYP1B1 protein expression:: A novel mechanism of anticancer drug resistance11Abbreviations: CYP

, Biochem. Pharmacol, vol.62, issue.2, pp.207-212, 2001.

U. M. Zanger and K. Klein, Pharmacogenetics of cytochrome P450 2B6 (CYP2B6): advances on polymorphisms, mechanisms, and clinical relevance, vol.4, 2013.

M. Goetz, A. Kamal, and M. Ames, « Tamoxifen Pharmacogenomics: The Role of CYP2D6 as a Predictor of Drug Response, Clin. Pharmacol. Ther, vol.83, issue.1, pp.160-166, 2008.

G. S. Hwang, R. Bhat, R. D. Crutchley, and M. V. Trivedi, Impact of CYP2D6 polymorphisms on endoxifen concentrations and breast cancer outcomes, 2017.

L. Dean, T. Therapy, ;. V. Genotype, H. Pratt, L. Mcleod et al., Éd. Bethesda (MD): National Center for Biotechnology Information (US), 2012.

V. J. Wacher, J. A. Silverman, Y. Zhang, and L. Z. Benet, « Role of P-glycoprotein and cytochrome P450 3A in limiting oral absorption of peptides and peptidomimetics, J. Pharm. Sci, vol.87, issue.11, pp.1322-1330, 1998.

I. Royer, B. Monsarrat, M. Sonnier, M. Wright, and T. Cresteil, Metabolism of docetaxel by human cytochromes P450: interactions with paclitaxel and other antineoplastic drugs, vol.56, pp.58-65, 1996.

Y. Miyoshi, A. Ando, Y. Takamura, T. Taguchi, Y. Tamaki et al., « Prediction of response to docetaxel by CYP3A4 mRNA expression in breast cancer tissues », Int. J. Cancer, vol.97, issue.1, pp.129-132, 2002.

C. Martínez, E. García-martín, R. M. Pizarro, F. J. García-gamito, and J. A. Agúndez, « Expression of paclitaxel-inactivating CYP3A activity in human colorectal cancer: implications for drug therapy », Br. J. Cancer, vol.87, issue.6, pp.681-686, 2002.

A. Beck, « A role for dihydropyrimidine dehydrogenase and thymidylate synthase in tumour sensitivity to fluorouracil », Eur. J. Cancer, vol.30, issue.10, pp.1517-1522, 1994.

M. E. Nita, O. Tominaga, H. Nagawa, T. Tsuruo, and T. Muto, « Dihydropyrimidine dehydrogenase but not thymidylate synthase expression is associated with resistance to 5-fluorouracil in colorectal cancer, », Hepatogastroenterology, vol.45, pp.2117-2122, 1997.

M. C. Etienne, « Response to fluorouracil therapy in cancer patients: the role of tumoral dihydropyrimidine dehydrogenase activity. », J. Clin. Oncol, vol.13, issue.7, pp.1663-1670, 1995.

D. Salonga, « Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase, Clin. Cancer Res, vol.6, issue.4, pp.1322-1327, 2000.

L. Dean, F. Therapy, ;. V. Genotype, H. Pratt, L. Mcleod et al., Éd. Bethesda (MD): National Center for Biotechnology Information (US), 2012.

L. Iyer, Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. », J. Clin. Invest, vol.101, p.847, 1998.

J. Cummings, « Glucuronidation as a Mechanism of Intrinsic Drug Resistance in Human Colon Cancer Reversal of Resistance by Food Additives, Cancer Res, vol.63, issue.23, pp.8443-8450, 2003.

T. Takahashi, Y. Fujiwara, M. Yamakido, O. Katoh, H. Watanabe et al., The Role of Glucuronidation in 7-Ethyl-10-hydroxycamptothecin Resistance in vitro, vol.88, pp.1211-1217, 1997.

D. Townsend, K. Tew, D. M. Townsend, and K. D. Tew, The role of glutathione-Stransferase in anti-cancer drug resistance, Oncogene, vol.22, pp.7369-7375, 2003.

M. M. Gottesman, T. Fojo, and S. E. Bates, « Multidrug resistance in cancer: role of ATP-dependent transporters », Nat. Rev. Cancer, vol.2, issue.1, pp.48-58, 2002.

D. R. Ferry, M. A. Russell, and M. H. Cullen, « P-glycoprotein possesses a 1, 4-dihydropyridine-selective drug acceptor site which is alloserically coupled to a vincaalkaloid-selective binding site, Biochem. Biophys. Res. Commun, vol.188, issue.1, pp.440-445, 1992.

M. M. Gottesman and I. Pastan, Biochemistry of multidrug resistance mediated by the multidrug transporter, vol.62, pp.385-427, 1993.

M. F. Rosenberg, R. Callaghan, R. C. Ford, and C. F. Higgins, « Structure of the multidrug resistance P-glycoprotein to 2.5 nm resolution determined by electron microscopy and image analysis, J. Biol. Chem, vol.272, pp.10685-10694, 1997.

C. M. Armstrong and A. C. Gao, « Drug resistance in castration resistant prostate cancer: resistance mechanisms and emerging treatment strategies, Am. J. Clin. Exp. Urol, vol.3, issue.2, p.64, 2015.

A. J. O'neill, « Characterisation and manipulation of docetaxel resistant prostate cancer cell lines, Mol. Cancer, vol.10, p.126, 2011.

Y. Zhu, « Inhibition of ABCB1 expression overcomes acquired docetaxel resistance in prostate cancer, Mol. Cancer Ther, vol.12, issue.9, p.1829, 2013.

Y. Zhu, C. Liu, C. Armstrong, W. Lou, A. Sandher et al., « Anti-androgens inhibit ABCB1 efflux and ATPase activity and reverse docetaxel resistance in advanced prostate cancer, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.21, p.4133, 2015.

P. M. Fracasso, « Phase I study of docetaxel in combination with the Pglycoprotein inhibitor, zosuquidar, in resistant malignancies, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.10, p.7220, 2004.

L. Van-zuylen, « Disposition of docetaxel in the presence of P-glycoprotein inhibition by intravenous administration of R101933 », Eur. J. Cancer Oxf. Engl, vol.38, issue.8, pp.1090-1099, 1990.

A. P. Lombard, « ABCB1 Mediates Cabazitaxel-Docetaxel Cross-Resistance in Advanced Prostate Cancer, Mol. Cancer Ther, 2017.

Y. Xie, « The 44-kDa Pim-1 Kinase Phosphorylates BCRP/ABCG2 and Thereby Promotes Its Multimerization and Drug-resistant Activity in Human Prostate Cancer Cells, J. Biol. Chem, vol.283, issue.6, pp.3349-3356, 2008.

N. A. Colabufo, « Bicalutamide failure in prostate cancer treatment: Involvement of Multi Drug Resistance proteins », Eur. J. Pharmacol, vol.601, issue.1, p.38, 2008.

K. Kim, Y. Cha, H. Lee, H. Joo, and J. Park, Effects of the ABCG2 and ABCB1 drug transporter polymorphisms on the pharmacokinetics of bicalutamide in humans, vol.438, pp.7-11, 2015.

T. M. Sissung, « ABCB1 genetic variation influences the toxicity and clinical outcome of patients with androgen independent prostate cancer treated with docetaxel, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.14, pp.4543-4549, 2008.

T. Hsia, C. Lin, J. Wang, S. Ho, and E. A. Kao, « Relationship between chemotherapy response of small cell lung cancer and P-glycoprotein or multidrug resistance-related protein expression, Lung, vol.180, issue.3, pp.173-179, 2002.

K. Nooter, « Expression of the multidrug resistance-associated protein (MRP) gene in primary non-small-cell lung cancer, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol, vol.7, issue.1, p.75, 1996.

, « MRP1 not MDR1 gene expression is the predominant mechanism of acquired multidrug resistance in two prostate carcinoma cell lines, vol.15, 2000.

. Doi101038sjpcan4500394, , vol.3, 2000.

M. J. Grzywacz, J. Yang, and W. N. Hait, Effect of the Multidrug Resistance Protein on the Transport of the Antiandrogen Flutamide, vol.63, pp.2492-2498, 2003.

P. Germain, B. Staels, C. Dacquet, M. Spedding, and V. Laudet, « Overview of nomenclature of nuclear receptors », Pharmacol. Rev, vol.58, issue.4, pp.685-704, 2006.

S. M. Hollenberg, « Primary structure and expression of a functional human glucocorticoid receptor cDNA, Nature, vol.318, pp.635-641, 1985.

M. Petkovich, N. J. Brand, A. Krust, and P. Chambon, « A human retinoic acid receptor which belongs to the family of nuclear receptors, Nature, vol.330, pp.444-450, 1987.

S. A. Kliewer, J. M. Lehmann, and T. M. Willson, « Orphan nuclear receptors: shifting endocrinology into reverse, Science, vol.284, pp.757-760, 1999.

Y. Li, M. H. Lambert, and H. E. Xu, Activation of nuclear receptors: a perspective from structural genomics », Structure, vol.11, pp.741-746, 2003.

A. Chawla, J. J. Repa, R. M. Evans, and D. J. Mangelsdorf, Nuclear receptors and lipid physiology: opening the X-files, vol.294, pp.1866-1870, 2001.

C. , Etude du rôle du récepteur des xénobiotiques PXR dans la réponse à la chimiothérapie du cancer colorectal, vol.1, 2009.

B. Blumberg, « BXR, an embryonic orphan nuclear receptor activated by a novel class of endogenous benzoate metabolites, Genes Dev, vol.12, issue.9, pp.1269-1277, 1998.

S. A. Kliewer, « An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway, Cell, vol.92, issue.1, pp.73-82, 1998.

B. Blumberg, SXR, a novel steroid and xenobioticsensing nuclear receptor, vol.12, pp.3195-3205, 1998.

G. Bertilsson, « Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction, Proc. Natl. Acad. Sci, vol.95, pp.12208-12213, 1998.

J. M. Lehmann, D. D. Mckee, M. A. Watson, T. M. Willson, J. T. Moore et al., « The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. », J. Clin. Invest, vol.102, issue.5, p.1016, 1998.

M. Baes, T. Gulick, H. S. Choi, M. G. Martinoli, D. Simha et al., « A new orphan member of the nuclear hormone receptor superfamily that interacts with a subset of retinoic acid response elements. », Mol. Cell. Biol, vol.14, issue.3, pp.1544-1552, 1994.

P. Honkakoski, I. Zelko, T. Sueyoshi, and M. Negishi, « The nuclear orphan receptor CAR-retinoid X receptor heterodimer activates the phenobarbital-responsive enhancer module of the CYP2B gene, Mol. Cell. Biol, vol.18, issue.10, pp.5652-5658, 1998.

Y. Wang, S. S. Ong, S. C. Chai, and E. T. Chen, Role of CAR and PXR in xenobiotic sensing and metabolism, Expert Opin. Drug Metab. Toxicol, vol.8, issue.7, p.803, 2012.

R. E. Watkins, « The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity, Science, vol.292, pp.2329-2333, 2001.

J. Orans, D. G. Teotico, and M. R. Redinbo, « The nuclear xenobiotic receptor pregnane X receptor: recent insights and new challenges, Mol. Endocrinol, vol.19, pp.2891-2900, 2005.

M. Saradhi, A. Sengupta, G. Mukhopadhyay, R. K. Tyagi, «. Pregnane et al., PXR/SXR) resides predominantly in the nuclear compartment of the interphase cell and associates with the condensed chromosomes during mitosis, Biochim. Biophys. Acta BBA-Mol. Cell Res, vol.1746, issue.2, pp.85-94, 2005.

H. Li and H. Wang, Activation of xenobiotic receptors: driving into the nucleus, Expert Opin. Drug Metab. Toxicol, vol.6, issue.4, pp.409-426, 2010.

D. D. Moore, « International Union of Pharmacology. LXII. The NR1H and NR1I receptors: constitutive androstane receptor, pregnene X receptor, farnesoid X receptor ?, farnesoid X receptor ?, liver X receptor ?, liver X receptor ?, and vitamin D receptor, Pharmacol. Rev, vol.58, issue.4, pp.742-759, 2006.

N. Scheer, « A novel panel of mouse models to evaluate the role of human pregnane X receptor and constitutive androstane receptor in drug response, J. Clin. Invest, vol.118, issue.9, pp.3228-3239, 2008.

J. Y. Cui, S. S. Gunewardena, C. E. Rockwell, and C. D. Klaassen, « ChIPing the cistrome of PXR in mouse liver, Nucleic Acids Res, p.654, 2010.

S. R. Faucette, T. Sueyoshi, C. M. Smith, M. Negishi, E. L. Lecluyse et al., « Differential regulation of hepatic CYP2B6 and CYP3A4 genes by constitutive androstane receptor but not pregnane X receptor, J. Pharmacol. Exp. Ther, vol.317, issue.3, pp.1200-1209, 2006.

W. Xie, « Reciprocal activation of xenobiotic response genes by nuclear receptors SXR/PXR and CAR, Genes Dev, vol.14, issue.23, pp.3014-3023, 2000.

B. Goodwin, E. Hodgson, D. J. Costa, G. R. Robertson, and C. Liddle, « Transcriptional regulation of the human CYP3A4gene by the constitutive androstane receptor, Mol. Pharmacol, vol.62, issue.2, pp.359-365, 2002.

C. Handschin and U. A. Meyer, « Induction of drug metabolism: the role of nuclear receptors », Pharmacol. Rev, vol.55, p.649, 2003.

A. H. Tolson and H. Wang, « Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR, Adv. Drug Deliv. Rev, vol.62, 2010.

N. Hariparsad, « Identification of pregnane-X receptor target genes and coactivator and corepressor binding to promoter elements in human hepatocytes », Nucleic Acids Res, p.1047, 2009.

H. Zaher, T. J. Yang, H. V. Gelboin, P. Fernandez-salguero, and F. J. Gonzalez, Effect of Phenobarbital on Hepatic CYP1A1 and CYP1A2 in the Ahr-Null Mouse, vol.55, p.235, 1998.

K. Yoshinari, N. Yoda, T. Toriyabe, and Y. Yamazoe, « Constitutive androstane receptor transcriptionally activates human CYP1A1 and CYP1A2 genes through a common regulatory element in the 5?-flanking region, Biochem. Pharmacol, vol.79, issue.2, pp.261-269, 2010.

J. Sugatani, « The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR, Hepatology, vol.33, issue.5, pp.1232-1238, 2001.

W. Xie, « Control of steroid, heme, and carcinogen metabolism by nuclear pregnane X receptor and constitutive androstane receptor, Proc. Natl. Acad. Sci. U. S. A, vol.100, issue.7, p.4150, 2003.

O. Burk, K. A. Arnold, A. Geick, H. Tegude, and M. Eichelbaum, « A role for constitutive androstane receptor in the regulation of human intestinal MDR1 expression », Biol. Chem, vol.386, issue.6, pp.503-513, 2005.

A. Geick, M. Eichelbaum, and O. Burk, « Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin, J. Biol. Chem, vol.276, pp.14581-14587, 2001.

H. R. Kast, « Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor, J. Biol. Chem, vol.277, issue.4, pp.2908-2915, 2002.

X. C. Kretschmer and W. S. Baldwin, « CAR and PXR: xenosensors of endocrine disrupters?, Chem. Biol. Interact, vol.155, issue.3, pp.111-128, 2005.

S. S. Ong, S. C. Chai, T. Chen, and Y. Wang, Pregnane X receptor in drug development, 2011.

J. L. Staudinger, « The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity, Proc. Natl. Acad. Sci, vol.98, pp.3369-3374, 2001.

W. Mnif, Estrogens and antiestrogens activate hPXR, vol.170, pp.19-29, 2007.

N. Landes, « Vitamin E activates gene expression via the pregnane X receptor, Biochem. Pharmacol, vol.65, issue.2, pp.269-273, 2003.

C. Zhou, M. M. Tabb, A. Sadatrafiei, F. Grün, and B. Blumberg, « Tocotrienols activate the steroid and xenobiotic receptor, SXR, and selectively regulate expression of its target genes, Drug Metab. Dispos, vol.32, issue.10, pp.1075-1082, 2004.

M. M. Tabb, « Vitamin K2 regulation of bone homeostasis is mediated by the steroid and xenobiotic receptor SXR, J. Biol. Chem, vol.278, pp.43919-43927, 2003.

D. Kluth, A. Banning, I. Paur, and R. Blomhoff, Brigelius-Flohé, « Modulation of pregnane X receptor-and electrophile responsive element-mediated gene expression by dietary polyphenolic compounds », Free Radic, Biol. Med, vol.42, issue.3, p.315, 2007.

L. B. Moore, John's wort induces hepatic drug metabolism through activation of the pregnane X receptor, Proc. Natl. Acad. Sci, vol.97, pp.7500-7502, 2000.

I. Dussault, M. Lin, K. Hollister, E. H. Wang, T. W. Synold et al., « Peptide mimetic HIV protease inhibitors are ligands for the orphan receptor SXR, J. Biol. Chem, vol.276, pp.33309-33312, 2001.

T. M. Willson, S. A. Kliewer, and «. Pxr, CAR and drug metabolism », Nat. Rev. Drug Discov, vol.1, pp.259-266, 2002.

S. Harmsen, I. Meijerman, R. F. Maas-bakker, J. H. Beijnen, and J. H. Schellens, « PXR-mediated P-glycoprotein induction by small molecule tyrosine kinase inhibitors », Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci, vol.48, issue.5, pp.644-649, 2013.

T. W. Synold, I. Dussault, and B. M. Forman, The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux, vol.7, pp.584-590, 2001.

C. Zhou, « The dietary isothiocyanate sulforaphane is an antagonist of the human steroid and xenobiotic nuclear receptor, Mol. Pharmacol, vol.71, issue.1, pp.220-229, 2007.

A. Takeshita, M. Taguchi, N. Koibuchi, and Y. Ozawa, « Putative role of the orphan nuclear receptor SXR (steroid and xenobiotic receptor) in the mechanism of CYP3A4 inhibition by xenobiotics, J. Biol. Chem, vol.277, pp.32453-32458, 2002.

H. Li, « Novel Yeast-based Strategy Unveils Antagonist Binding Regions on the Nuclear Xenobiotic Receptor PXR, J. Biol. Chem, vol.288, p.13655, 2013.

I. Fuchs, « Effect of the CYP3A inhibitor ketoconazole on the PXR-mediated induction of CYP3A activity, Eur. J. Clin. Pharmacol, vol.69, issue.3, pp.507-513, 2013.

B. C. Das, « Synthesis of novel ketoconazole derivatives as inhibitors of the human Pregnane X Receptor (PXR; NR1I2; also termed SXR, PAR) », Bioorg. Med. Chem. Lett, vol.18, p.3974, 2008.

M. Venkatesh, « In Vivo and In Vitro Characterization of a First-in-Class Novel Azole Analog That Targets Pregnane X Receptor Activation, Mol. Pharmacol, vol.80, issue.1, p.124, 2011.

S. C. Chai, M. T. Cherian, Y. Wang, and T. Chen, « Small-molecule modulators of PXR and CAR, Biochim. Biophys. Acta, vol.1859, issue.9, p.1141, 2016.

S. Mani, W. Dou, and M. R. Redinbo, « PXR antagonists and implication in drug metabolism, Drug Metab. Rev, vol.45, issue.1, pp.60-72, 2013.

C. Zhou, N. King, K. Y. Chen, and J. L. Breslow, « Activation of PXR induces hypercholesterolemia in wild-type and accelerates atherosclerosis in apoE deficient mice, J. Lipid Res, vol.50, issue.10, pp.2004-2013, 2009.

D. Lütjohann, « Influence of rifampin on serum markers of cholesterol and bile acid synthesis in men. », Int. J. Clin. Pharmacol. Ther, vol.42, issue.6, pp.307-313, 2004.

J. M. Eiris, « Effects of long-term treatment with antiepileptic drugs on serum lipid levels in children with epilepsy, Neurology, vol.45, issue.6, pp.1155-1157, 1995.

S. P. Saini, « A novel constitutive androstane receptor-mediated and CYP3A-independent pathway of bile acid detoxification, Mol. Pharmacol, vol.65, issue.2, pp.292-300, 2004.

J. Sonoda, W. Xie, J. M. Rosenfeld, J. L. Barwick, P. S. Guzelian et al., « Regulation of a xenobiotic sulfonation cascade by nuclear pregnane X receptor (PXR), Proc. Natl. Acad. Sci, vol.99, pp.13801-13806, 2002.

M. Wagner, « CAR and PXR agonists stimulate hepatic bile acid and bilirubin detoxification and elimination pathways in mice, Hepatology, vol.42, issue.2, pp.420-430, 2005.

Y. Zhai, H. V. Pai, J. Zhou, J. A. Amico, R. R. Vollmer et al., Activation of pregnane X receptor disrupts glucocorticoid and mineralocorticoid homeostasis », Mol. Endocrinol, vol.21, issue.1, pp.138-147, 2007.

S. Kodama, R. Moore, Y. Yamamoto, and M. Negishi, « Human nuclear pregnane X receptor cross-talk with CREB to repress cAMP activation of the glucose-6-phosphatase gene », Biochem. J, vol.407, issue.3, pp.373-381, 2007.

Y. Konno, M. Negishi, and E. S. Kodama, « The roles of nuclear receptors CAR and PXR in hepatic energy metabolism, Drug Metab. Pharmacokinet, vol.23, issue.1, pp.8-13, 2008.

Y. Uno, « Characterization of six base pair deletion in the putative HNF1-binding site of human PXR promoter, J. Hum. Genet, vol.48, issue.11, pp.594-597, 2003.

J. Lamba, V. Lamba, S. Strom, R. Venkataramanan, and E. E. Schuetz, « Novel single nucleotide polymorphisms in the promoter and intron 1 of human pregnane X receptor/NR1I2 and their association with CYP3A4 expression, Drug Metab. Dispos, vol.36, issue.1, pp.169-181, 2008.

R. D. Patel, B. D. Hollingshead, C. J. Omiecinski, and G. H. Perdew, « Arylhydrocarbon receptor activation regulates constitutive androstane receptor levels in murine and human liver, Hepatology, vol.46, issue.1, pp.209-218, 2007.

N. Iwazaki, « Involvement of hepatocyte nuclear factor 4alpha in transcriptional regulation of the human pregnane X receptor gene in the human liver, Drug Metab. Pharmacokinet, vol.23, issue.1, pp.59-66, 2008.

J. M. Pascussi, « Differential regulation of constitutive androstane receptor expression by hepatocyte nuclear factor4? isoforms, Hepatology, vol.45, issue.5, pp.1146-1153, 2007.

A. Misawa, « Methylation-associated silencing of the nuclear receptor 1I2 gene in advanced-type neuroblastomas, identified by bacterial artificial chromosome arraybased methylated CpG island amplification, Cancer Res, vol.65, pp.10233-10242, 2005.

W. Habano, « Involvement of promoter methylation in the regulation of Pregnane X receptor in colon cancer cells, BMC Cancer, vol.11, p.81, 2011.

M. Yang and J. Y. Park, « DNA methylation in promoter region as biomarkers in prostate cancer », Methods Mol. Biol. Clifton NJ, vol.863, pp.67-109, 2012.

S. Takagi, M. Nakajima, T. Mohri, and T. Yokoi, « Post-transcriptional regulation of human pregnane X receptor by micro-RNA affects the expression of cytochrome P450 3A4, J. Biol. Chem, vol.283, pp.9674-9680, 2008.

Y. Fujita, « MiR-148a Attenuates Paclitaxel Resistance of Hormone-refractory, Drug-resistant Prostate Cancer PC3 Cells by Regulating MSK1 Expression, J. Biol. Chem, vol.285, 2010.

S. R. Pondugula, C. Brimer-cline, J. Wu, E. G. Schuetz, R. K. Tyagi et al., « A phosphomimetic mutation at threonine-57 abolishes transactivation activity and alters nuclear localization pattern of human pregnane x receptor, Drug Metab. Dispos, vol.37, issue.4, pp.719-730, 2009.

X. Ding and J. L. Staudinger, « Repression of PXR-mediated induction of hepatic CYP3A gene expression by protein kinase C », Biochem. Pharmacol, vol.69, issue.5, pp.867-873, 2005.

H. Masuyama, H. Inoshita, Y. Hiramatsu, and T. Kudo, « Ligands have various potential effects on the degradation of pregnane X receptor by proteasome, Endocrinology, vol.143, issue.1, pp.55-61, 2002.

H. Masuyama, N. Suwaki, Y. Tateishi, H. Nakatsukasa, T. Segawa et al., « The pregnane X receptor regulates gene expression in a ligand-and promoter-selective fashion, Mol. Endocrinol, vol.19, issue.5, pp.1170-1180, 2005.

W. Cui, M. Sun, N. Galeva, T. D. Williams, Y. Azuma et al., SUMOylation and Ubiquitylation Circuitry Controls Pregnane X Receptor Biology in Hepatocytes, vol.43, pp.1316-1325, 2015.

D. , « Acetylation of lysine 109 modulates pregnane X receptor DNA binding and transcriptional activity, Biochim. Biophys. Acta, vol.1859, issue.9, pp.1155-1169, 2016.

S. R. Pondugula, P. Pavek, S. Mani, and . Pregnane, Receptor and Cancer: Context-Specificity is Key », Nucl. Recept. Res, vol.3, 2016.

Y. Niu, « Activated pregnane X receptor inhibits cervical cancer cell proliferation and tumorigenicity by inducing G2/M cell-cycle arrest, Cancer Lett, vol.347, issue.1, pp.88-97, 2014.

N. Ouyang, « Pregnane X receptor suppresses proliferation and tumourigenicity of colon cancer cells, Br. J. Cancer, vol.102, pp.1753-1761, 2010.

H. Wang, « Pregnane X receptor activation induces FGF19-dependent tumor aggressiveness in humans and mice, J. Clin. Invest, vol.121, issue.8, p.3220, 2011.

Y. Chen, Y. Tang, S. Chen, and D. Nie, « Regulation of drug resistance by human pregnane X receptor in breast cancer, Cancer Biol. Ther, vol.8, issue.13, p.1265, 2009.

E. Qiao and H. Yang, « Effect of pregnane X receptor expression on drug resistance in breast cancer, Oncol. Lett, vol.7, p.1191, 2014.

C. , « Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation, Mol. Cancer, vol.9, p.46, 2010.

A. Basseville, « Irinotecan induces steroid and xenobiotic receptor (SXR) signaling to detoxification pathway in colon cancer cells, Mol. Cancer, vol.10, p.80, 2011.

S. Harmsen, I. Meijerman, J. H. Beijnen, and J. H. Schellens, « Nuclear receptor mediated induction of cytochrome P450 3A4 by anticancer drugs: a key role for the pregnane X receptor, Cancer Chemother. Pharmacol, vol.64, issue.1, pp.35-43, 2009.

S. Harmsen, I. Meijerman, C. L. Febus, R. F. Maas-bakker, J. H. Beijnen et al., « PXR-mediated induction of P-glycoprotein by anticancer drugs in a human colon adenocarcinoma-derived cell line, Cancer Chemother. Pharmacol, vol.66, issue.4, pp.765-771, 2010.

H. Guchelaar, C. H. Napel, E. G. De-vries, and N. H. Mulder, « Clinical, toxicological and pharmaceutical aspects of the antineoplastic drug taxol: A review, Clin. Oncol, vol.6, issue.1, p.40, 1994.

R. Roskoski, « A historical overview of protein kinases and their targeted small molecule inhibitors », Pharmacol. Res, vol.100, pp.1-23, 2015.

J. Merlin, « Les inhibiteurs de tyrosine kinase en oncologie, Lett. Pharmacol, vol.22, issue.2, pp.51-62, 2008.

D. A. Walsh, J. P. Perkins, and E. G. Krebs, « An adenosine 3',5'-monophosphatedependant protein kinase from rabbit skeletal muscle, J. Biol. Chem, vol.243, p.3763, 1968.

P. Cohen, The origins of protein phosphorylation, Nat. Cell Biol, vol.4, issue.5, pp.127-130, 2002.

C. Neul, E. Schaeffeler, A. Sparreboom, S. Laufer, M. Schwab et al., « Impact of Membrane Drug Transporters on Resistance to Small-Molecule Tyrosine Kinase Inhibitors, Trends Pharmacol. Sci, vol.37, issue.11, p.904, 2016.

G. Manning, D. B. Whyte, R. Martinez, T. Hunter, and S. Sudarsanam, « The protein kinase complement of the human genome, Science, vol.298, 2002.

D. R. Knighton, « Crystal structure of the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase, Science, vol.253, pp.407-414, 1991.

S. Boutayeb, Pathol. Biol, vol.60, pp.229-233, 2012.

B. W. Murray and N. Miller, « Durability of Kinase-Directed Therapies--A Network Perspective on Response and Resistance, Mol. Cancer Ther, vol.14, issue.9, 2015.

M. Juchum, M. Günther, and S. A. Laufer, « Fighting cancer drug resistance: Opportunities and challenges for mutation-specific EGFR inhibitors, Drug Resist. Updat. Rev. Comment. Antimicrob. Anticancer Chemother, vol.20, pp.12-28, 2015.

S. Soverini, Contribution of ABL Kinase Domain Mutations to Imatinib Resistance in Different Subsets of Philadelphia-Positive Patients: By the GIMEMA Working Party on Chronic Myeloid Leukemia, vol.12, pp.7374-7379, 2006.

N. Wagle, « Dissecting Therapeutic Resistance to RAF Inhibition in Melanoma by Tumor Genomic Profiling, J. Clin. Oncol, vol.29, pp.3085-3096, 2011.

M. K. Callahan, « Progression of RAS-Mutant Leukemia during RAF Inhibitor Treatment, N. Engl. J. Med, vol.367, pp.2316-2321

P. Wu, T. E. Nielsen, and M. H. Clausen, « FDA-approved small-molecule kinase inhibitors, Trends Pharmacol. Sci, vol.36, issue.7, pp.422-439, 2015.

M. Baccarani, F. Castagnetti, G. Gugliotta, and G. Rosti, « A review of the European LeukemiaNet recommendations for the management of CML, Ann. Hematol, vol.94, issue.2, pp.141-147, 2015.

Z. Eroglu and A. Ribas, « Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and place in therapy, Ther. Adv. Med. Oncol, vol.8, issue.1, p.48, 2016.

A. Gater, « Adherence to oral tyrosine kinase inhibitor therapies in chronic myeloid leukemia, Leuk. Res, vol.36, issue.7, p.817, 2012.

L. Wood, « A review on adherence management in patients on oral cancer therapies, Eur. J. Oncol. Nurs, vol.16, p.432, 2012.

S. Antoun, V. E. Baracos, L. Birdsell, B. Escudier, and M. B. Sawyer, « Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma, Ann. Oncol, vol.21, issue.8, pp.1594-1598, 2010.

O. Huillard, « Sarcopenia and body mass index predict sunitinib-induced early dose-limiting toxicities in renal cancer patients », Br. J. Cancer, vol.108, issue.5, pp.1034-1041, 2013.

M. H. Diekstra, « CYP3A5 and ABCB1 polymorphisms as predictors for sunitinib outcome in metastatic renal cell carcinoma, Eur. Urol, vol.68, pp.621-629, 2015.

P. Boudou-rouquette, « Early sorafenib-induced toxicity is associated with drug exposure and UGTIA9 genetic polymorphism in patients with solid tumors: a preliminary study, PloS One, vol.7, issue.8, p.42875, 2012.

N. Thatcher, « Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebocontrolled, multicentre study (Iressa Survival Evaluation in Lung Cancer), Lancet Lond. Engl, vol.366, pp.1527-1537, 2005.

H. Klümpen, C. F. Samer, R. H. Mathijssen, J. H. Schellens, and H. Gurney, « Moving towards dose individualization of tyrosine kinase inhibitors, Cancer Treat. Rev, vol.37, issue.4, pp.251-260, 2011.

D. R. Duckett and M. D. Cameron, « Metabolism considerations for kinase inhibitors in cancer treatment, Expert Opin. Drug Metab. Toxicol, vol.6, issue.10, pp.1175-1193, 2010.

T. Illmer, « P-glycoprotein-mediated drug efflux is a resistance mechanism of chronic myelogenous leukemia cells to treatment with imatinib mesylate, Leukemia, vol.18, issue.3, pp.401-408, 2004.

J. Thomas, L. Wang, R. E. Clark, and M. Pirmohamed, « Active transport of imatinib into and out of cells: implications for drug resistance, Blood, vol.104, p.3739, 2004.

C. Heged?s, « Interaction of nilotinib, dasatinib and bosutinib with ABCB1 and ABCG2: implications for altered anti-cancer effects and pharmacological properties: Interaction of Bcr-Abl inhibitors and MDR-ABC proteins », Br. J. Pharmacol, vol.158, issue.4, pp.1153-1164, 2009.

J. Deng, J. Shao, J. S. Markowitz, and E. G. An, « ABC Transporters in Multi-Drug Resistance and ADME-Tox of Small Molecule Tyrosine Kinase Inhibitors », Pharm. Res, vol.31, issue.9, pp.2237-2255, 2014.

C. E. Geyer, « Lapatinib plus Capecitabine for HER2-Positive Advanced Breast Cancer, N. Engl. J. Med, vol.355, pp.2733-2743, 2006.

K. Yanase, S. Tsukahara, S. Asada, E. Ishikawa, Y. Imai et al., « Gefitinib reverses breast cancer resistance protein-mediated drug resistance, Mol. Cancer Ther, vol.3, issue.9, pp.1119-1125, 2004.

Z. Shi, OSI-774) antagonizes ATP-binding cassette subfamily B member 1 and ATP-binding cassette subfamily G member 2-mediated drug resistance, Cancer Res, vol.67, p.11012, 2007.

D. K. Hiwase, D. White, S. Zrim, V. Saunders, J. V. Melo et al., « Nilotinib-mediated inhibition of ABCB1 increases intracellular concentration of dasatinib in CML cells: implications for combination TKI therapy, Leukemia, vol.24, issue.3, p.660, 2010.

D. L. White, V. A. Saunders, S. R. Quinn, P. W. Manley, and T. P. Hughes, « Imatinib increases the intracellular concentration of nilotinib, which may explain the observed synergy between these drugs, Blood, vol.109, issue.8, p.3609, 2007.

H. Burger, « Chronic imatinib mesylate exposure leads to reduced intracellular drug accumulation by induction of the ABCG2 (BCRP) and ABCB1 (MDR1) drug transport pumps, Cancer Biol. Ther, vol.4, issue.7, p.747, 2005.

N. Anreddy, P. Gupta, R. J. Kathawala, A. Patel, J. N. Wurpel et al., « Tyrosine kinase inhibitors as reversal agents for ABC transporter mediated drug resistance, Mol. Basel Switz, vol.19, issue.9, pp.13848-13877, 2014.

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, issue.1, pp.16-23, 1979.

K. R. Stone, D. D. Mickey, H. Wunderli, G. H. Mickey, and D. F. Paulson, « Isolation of a human prostate carcinoma cell line (DU 145) », Int. J. Cancer, vol.21, issue.3, pp.274-281, 1978.

J. Veldscholte, « A mutation in the ligand binding domain of the androgen receptor of human INCaP cells affects steroid binding characteristics and response to antiandrogens, Biochem. Biophys. Res. Commun, vol.173, issue.2, pp.534-540, 1990.

A. Van-bokhoven, « Molecular characterization of human prostate carcinoma cell lines, The Prostate, vol.57, pp.205-225, 2003.

Y. Li, S. C. Chan, L. J. Brand, T. H. Hwang, K. A. Silverstein et al., « Androgen receptor splice variants mediate enzalutamide resistance in castrationresistant prostate cancer cell lines, Cancer Res, vol.73, issue.2, pp.483-489, 2013.

J. Lehár, « Synergistic drug combinations tend to improve therapeutically relevant selectivity, Nat. Biotechnol, vol.27, issue.7, pp.659-666, 2009.

T. Fujimura, « Expression of cytochrome P450 3A4 and its clinical significance in human prostate cancer, Urology, vol.74, issue.2, pp.391-397, 2009.

M. Marion, O. Hantz, and D. Durantel, « The HepaRG cell line: biological properties and relevance as a tool for cell biology, drug metabolism, and virology studies », Methods Mol. Biol. Clifton NJ, vol.640, pp.261-272, 2010.

B. Rochat, J. M. Morsman, G. I. Murray, W. D. Figg, and H. L. Mcleod, « Human CYP1B1 and anticancer agent metabolism: mechanism for tumor-specific drug inactivation?, J. Pharmacol. Exp. Ther, vol.296, issue.2, pp.537-541, 2001.

G. G. Gibson, A. Phillips, S. Aouabdi, K. Plant, and N. Plant, « Transcriptional regulation of the human pregnane-X receptor, Drug Metab. Rev, vol.38, issue.2, pp.31-49, 2006.

S. Kumar, B. Jaiswal, S. Kumar, S. Negi, and R. K. Tyagi, « Cross-talk between androgen receptor and pregnane and xenobiotic receptor reveals existence of a novel modulatory action of anti-androgenic drugs, Biochem. Pharmacol, vol.80, issue.7, pp.964-976, 2010.

A. K. Macleod, L. A. Mclaughlin, C. J. Henderson, and C. R. Wolf, « Activation status of the pregnane X receptor influences vemurafenib availability in humanized mouse models, Cancer Res, vol.75, pp.4573-4581, 2015.

G. Gravis, « Results from a monocentric phase II trial of erlotinib in patients with metastatic prostate cancer, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO, vol.19, issue.9, pp.1624-1628, 2008.

J. B. Aragon-ching, « Final Analysis Of A Phase II Trial Using Sorafenib For Metastatic Castration Resistant Prostate Cancer, BJU Int, vol.103, p.1636, 2009.

M. L. Gonzalgo, C. P. Pavlovich, S. M. Lee, and W. G. Nelson, « Prostate cancer detection by GSTP1 methylation analysis of postbiopsy urine specimens, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.9, issue.7, pp.2673-2677, 2003.

G. J. Van-steenbrugge, C. J. Van-uffelen, J. Bolt, and F. H. Schröder, « The human prostatic cancer cell line LNCaP and its derived sublines: an in vitro model for the study of androgen sensitivity, J. Steroid Biochem. Mol. Biol, vol.40, pp.207-214, 1991.

P. Saxena, « PSA regulates androgen receptor expression in prostate cancer cells, The Prostate, vol.72, pp.769-776, 2012.

J. F. Deeken, J. H. Beumer, N. M. Anders, T. Wanjiku, M. Rusnak et al., « Preclinical assessment of the interactions between the antiretroviral drugs, ritonavir and efavirenz, and the tyrosine kinase inhibitor erlotinib, Cancer Chemother. Pharmacol, vol.76, issue.4, p.813, 2015.

H. Ellens, M. Johnson, S. K. Lawrence, C. Watson, L. Chen et al., Prediction of the Transporter-Mediated Drug-Drug Interaction Potential of Dabrafenib and Its Major Circulating Metabolites, vol.45, pp.646-656, 2017.

S. K. Lawrence, D. Nguyen, C. Bowen, L. Richards-peterson, and K. W. Skordos, « The metabolic drug-drug interaction profile of Dabrafenib: in vitro investigations and quantitative extrapolation of the P450-mediated DDI risk, Drug Metab. Dispos. Biol. Fate Chem, vol.42, issue.7, pp.1180-1190, 2014.

A. Hamada, « Effect of SLCO1B3 haplotype on testosterone transport and clinical outcome in caucasian patients with androgen-independent prostatic cancer, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.14, issue.11, pp.3312-3318, 2008.

R. A. Johnston, T. Rawling, T. Chan, F. Zhou, and E. M. Murray, « Selective inhibition of human solute carrier transporters by multikinase inhibitors, Drug Metab. Dispos. Biol. Fate Chem, vol.42, issue.11, pp.1851-1857, 2014.

M. Karlgren, « Classification of Inhibitors of Hepatic Organic Anion Transporting Polypeptides (OATPs): Influence of Protein Expression on Drug-Drug Interactions, J. Med. Chem, vol.55, issue.10, pp.4740-4763, 2012.

S. Wang, « Afatinib reverses multidrug resistance in ovarian cancer via dually inhibiting ATP binding cassette subfamily B member 1, Oncotarget, vol.6, pp.26142-26160, 2015.

E. Speir and . Cytomegalovirus, Gene Regulation by Reactive Oxygen Species: Agents in Atherosclerosis, Ann. N. Y. Acad. Sci, vol.899, issue.1, p.363, 2000.

P. Corazao-rozas, « Mitochondrial oxidative stress is the Achille's heel of melanoma cells resistant to Braf-mutant inhibitor, Oncotarget, vol.4, issue.11, 2013.

C. Planque, « Pregnane X-receptor promotes stem cell-mediated colon cancer relapse, Oncotarget, vol.7, p.56558, 2016.

R. K. Mittapalli, S. Vaidhyanathan, A. Z. Dudek, and W. F. Elmquist, « Mechanisms Limiting Distribution of the Threonine-Protein Kinase B-RaFV600E Inhibitor Dabrafenib to the Brain: Implications for the Treatment of Melanoma Brain Metastases, J. Pharmacol. Exp. Ther, vol.344, issue.3, pp.655-664, 2013.

S. Vaidhyanathan, R. K. Mittapalli, J. N. Sarkaria, and W. F. Elmquist, « Factors Influencing the CNS Distribution of a Novel MEK-1/2 Inhibitor: Implications for Combination Therapy for Melanoma Brain Metastases, Drug Metab. Dispos, vol.42, issue.8, pp.1292-1300, 2014.

J. Qiu, « Trametinib modulates cancer multidrug resistance by targeting ABCB1 transporter, Oncotarget, vol.6, pp.15494-15509, 2015.

N. R. Budha, « Evaluation of Cytochrome P450 3A4-Mediated Drug-Drug Interaction Potential for Cobimetinib Using Physiologically Based Pharmacokinetic Modeling and Simulation, Clin. Pharmacokinet, vol.55, issue.11, pp.1435-1445, 2016.