«. La-variable and . Ht-adjuvante, n'a finalement pas été retenue puisque l'HT adjuvante ne concerne que les cancers du sein et non l'ensemble des 3 cancers étudiés

. Le-seuil-de, 3 ng/mL était significativement associé à un risque de survenue d'une toxicité environ 4 fois plus important lorsque la C 0 est supérieure à 26, pp.95-143

C. Ng-/-ml,-«-nombre-de-ligne-de, «. Tc-», ». Présence-de-bisphosphonates, «. , ». et al., Analyse de Cox univariée Les variables « C 0 d'EVR binarisée » selon le seuil de 11

. Le-seuil-de, 9 ng/mL était significativement associé à une progression tumorale avec un risque 3,2 fois plus important lorsque la C 0 est inférieure à 11, pp.95-128

C. Le-nombre-de-ligne-de, TC était associé à un risque de progression tumorale 1,7 fois plus important

F. Saint-marcoux and F. Libert, ??l??ments de l???interpr??tation et du dialogue clinico-biologique pour quelques param??tres usuels de pharmacologie-toxicologie, Th??rapie, vol.71, issue.4, pp.405-434, 2016.
DOI : 10.1016/j.therap.2016.01.007

V. H. Mabasa and M. H. Ensom, The Role of Therapeutic Monitoring of Everolimus in Solid Organ Transplantation, Therapeutic Drug Monitoring, vol.27, issue.5, pp.666-676, 2005.
DOI : 10.1097/01.ftd.0000175911.70172.2e

R. C. Starling, Therapeutic Drug Monitoring for Everolimus in Heart Transplant Recipients Based on Exposure-Effect Modeling, American Journal of Transplantation, vol.3, issue.5, pp.2126-2131, 2004.
DOI : 10.1056/NEJMoa021744

J. M. Kovarik, Everolimus Therapeutic Concentration Range Defined from a Prospective Trial with Reduced-Exposure Cyclosporine in De Novo Kidney Transplantation, Therapeutic Drug Monitoring, vol.26, issue.5
DOI : 10.1097/00007691-200410000-00007

J. M. Kovarik, Exposure-response relationships for everolimus in de novo kidney transplantation: defining a therapeutic range, Transplantation, vol.73, issue.6, pp.920-925, 2002.
DOI : 10.1097/00007890-200203270-00016

M. Shipkova, Therapeutic Drug Monitoring of Everolimus, Therapeutic Drug Monitoring, vol.38, issue.2, pp.143-169, 2016.
DOI : 10.1097/FTD.0000000000000260

A. Ravaud, Relationship between everolimus exposure and safety and efficacy: Meta-analysis of clinical trials in oncology, European Journal of Cancer, vol.50, issue.3, pp.486-495, 1990.
DOI : 10.1016/j.ejca.2013.11.022

D. M. Sabatini, H. Erdjument-bromage, M. Lui, P. Tempst, and S. H. Snyder, RAFT1: A mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs, Cell, vol.78, issue.1, pp.35-43, 1994.
DOI : 10.1016/0092-8674(94)90570-3

E. J. Brown, A mammalian protein targeted by G1-arresting rapamycin???receptor complex, Nature, vol.369, issue.6483, pp.756-758, 1994.
DOI : 10.1038/369756a0

J. Baselga, Everolimus in Postmenopausal Hormone-Receptor???Positive Advanced Breast Cancer, New England Journal of Medicine, vol.366, issue.6, pp.520-529, 2012.
DOI : 10.1056/NEJMoa1109653

R. J. Motzer, Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial, The Lancet, vol.372, issue.9637, pp.449-456, 2008.
DOI : 10.1016/S0140-6736(08)61039-9

M. E. Pavel, Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study, The Lancet, vol.378, issue.9808, pp.2005-2012, 2011.
DOI : 10.1016/S0140-6736(11)61742-X

D. A. Yardley, Everolimus Plus Exemestane in Postmenopausal Patients with HR+ Breast Cancer: BOLERO-2 Final Progression-Free Survival Analysis, Advances in Therapy, vol.182, issue.3, pp.870-884, 2013.
DOI : 10.1007/s12325-013-0060-1

S. Dhillon, Everolimus in Combination with Exemestane: A Review of its Use in the Treatment of Patients with Postmenopausal Hormone Receptor-Positive, HER2-Negative Advanced Breast Cancer, Drugs, vol.28, issue.30, pp.475-485, 2013.
DOI : 10.1007/s40265-013-0034-2

F. Panzuto, Real-world study of everolimus in advanced progressive neuroendocrine tumors, The Oncologist, vol.20, issue.570, 2015.

M. Aapro, Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer, Annals of Oncology, vol.25, issue.4, pp.763-773, 2014.
DOI : 10.1093/annonc/mdu021

L. Albiges, Everolimus for patients with metastatic renal cell carcinoma refractory to anti-VEGF therapy: Results of a pooled analysis of non-interventional studies, European Journal of Cancer, vol.51, issue.16
DOI : 10.1016/j.ejca.2015.07.030

H. S. Rugo, Incidence and time course of everolimus-related adverse events in postmenopausal women with hormone receptor-positive advanced breast cancer: insights from BOLERO-2, Annals of Oncology, vol.25, issue.4, pp.808-815, 2014.
DOI : 10.1093/annonc/mdu009

D. Allorge and M. Loriot, Pharmacogenetics or the promise of a personalized medicine: variability in drug metabolism and transport], Ann. Biol. Clin. (Paris), vol.62, pp.499-511, 2004.

L. De-chaisemartin and M. Loriot, Pharmacog??n??tique des m??dicaments anticanc??reux, Pathologie Biologie, vol.53, issue.2, pp.116-124, 2005.
DOI : 10.1016/j.patbio.2004.05.001

N. Picard, K. Rouguieg-malki, N. Kamar, L. Rostaing, and P. Marquet, CYP3A5 Genotype Does Not Influence Everolimus In Vitro Metabolism and Clinical Pharmacokinetics in Renal Transplant Recipients, Transplantation, vol.91, issue.6, pp.652-656, 2011.
DOI : 10.1097/TP.0b013e31820ae4ac

URL : https://hal.archives-ouvertes.fr/inserm-00542366

D. Wang, Y. Guo, S. A. Wrighton, G. E. Cooke, and W. Sadee, Intronic polymorphism in CYP3A4 affects hepatic expression and response to statin drugs, The Pharmacogenomics Journal, vol.34, issue.4, pp.274-286, 2011.
DOI : 10.2217/pgs.09.42

L. Elens, polymorphism on calcineurin inhibitors??? dose requirements and trough blood levels in stable renal transplant patients, Pharmacogenomics, vol.12, issue.10, pp.1383-1396, 2011.
DOI : 10.2217/pgs.11.90

L. Elens, Novel CYP3A4 intron 6 single nucleotide polymorphism is associated with simvastatin-mediated cholesterol reduction in The Rotterdam Study, Pharmacogenetics and Genomics, vol.21, issue.12, pp.861-866, 2011.
DOI : 10.1097/FPC.0b013e32834c6edb

P. Kuehl, Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression, Nature Genetics, vol.27, issue.4, pp.383-391, 2001.
DOI : 10.1038/86882

M. Wong, CYP3A5 genotype and midazolam clearance in Australian patients receiving chemotherapy*1, Clinical Pharmacology & Therapeutics, vol.75, issue.6, pp.529-538, 2004.
DOI : 10.1016/j.clpt.2004.02.005

V. Haufroid, The effect of CYP3A5 and MDR1 (ABCB1) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood levels in stable renal transplant patients, Pharmacogenetics, vol.14, issue.3, pp.147-154, 2004.
DOI : 10.1097/00008571-200403000-00002

F. Josephson, CYP3A5 Genotype has an Impact on the Metabolism of the HIV Protease Inhibitor Saquinavir, Clinical Pharmacology & Therapeutics, vol.784, issue.5, pp.708-712, 2007.
DOI : 10.1038/sj.clpt.6100117

W. Jacobsen, Comparison of the in vitro metabolism of the macrolide immunosuppressants sirolimus and RAD, Transplantation Proceedings, vol.33, issue.1-2, pp.514-515, 2001.
DOI : 10.1016/S0041-1345(00)02116-3

R. J. Motzer, Phase 3 trial of everolimus for metastatic renal cell carcinoma, Cancer, vol.356, issue.suppl, pp.4256-4265, 2010.
DOI : 10.1002/cncr.25219

F. Lemaitre, Population Pharmacokinetics of Everolimus in Cardiac Recipients, Therapeutic Drug Monitoring, vol.34, issue.6, pp.686-694, 2012.
DOI : 10.1097/FTD.0b013e318273c899

D. J. Moes, Population Pharmacokinetics and Pharmacogenetics of Everolimus in Renal Transplant Patients, Clinical Pharmacokinetics, vol.13, issue.3, pp.467-480, 2012.
DOI : 10.2165/11599710-000000000-00000

K. E. Schoeppler, C. L. Aquilante, T. H. Kiser, D. N. Fish, and M. R. Zamora, The impact of genetic polymorphisms, diltiazem, and demographic variables on everolimus trough concentrations in lung transplant recipients, Clinical Transplantation, vol.11, issue.5, pp.590-597, 2014.
DOI : 10.1111/ctr.12350

D. J. Moes, Effect of CYP3A4*22, CYP3A5*3, and CYP3A Combined Genotypes on Cyclosporine, Everolimus, and Tacrolimus Pharmacokinetics in Renal Transplantation, CPT: Pharmacometrics Systems Pharmacology, vol.64, issue.2, p.100, 2014.
DOI : 10.1016/j.cmpb.2010.04.018

D. Kniepeiss, The role of CYP3A5 genotypes in dose requirements of tacrolimus and everolimus after heart transplantation, Clinical Transplantation, vol.8, issue.1, pp.146-150, 2011.
DOI : 10.1111/j.1399-0012.2009.01198.x

M. Neely and R. Jelliffe, Practical, Individualized Dosing: 21st Century Therapeutics and the Clinical Pharmacometrician, The Journal of Clinical Pharmacology, vol.48, issue.7, pp.842-847, 2010.
DOI : 10.1177/0091270009356572

F. Shihab, U. Christians, L. Smith, J. R. Wellen, and B. Kaplan, Focus on mTOR inhibitors and tacrolimus in renal transplantation: Pharmacokinetics, exposure???response relationships, and clinical outcomes, Transplant Immunology, vol.31, issue.1, pp.22-32, 2014.
DOI : 10.1016/j.trim.2014.05.002

J. M. Kovarik, Effect of Food on Everolimus Absorption: Quantification in Healthy Subjects and a Confirmatory Screening in Patients with Renal Transplants, Pharmacotherapy, vol.39, issue.2, pp.154-159, 2002.
DOI : 10.1023/A:1011940108365