F. Lamoureux and T. Duflot, , 2017.

, Pharmacogenetics in cardiovascular diseases: State of the art and implementationrecommendations of the French National Network of, Pharmacogenetics

, Therapie, vol.72, issue.2, pp.257-67

G. P. Aithal, C. P. Day, P. J. Kesteven, and A. K. Daly, Association of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications, Lancet, vol.353, issue.9154, pp.717-726, 1999.

M. J. Rieder, A. P. Reiner, B. F. Gage, D. A. Nickerson, C. S. Eby et al., Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose, N Engl J Med, vol.352, issue.22, pp.2285-93, 2005.

A. R. Shuldiner, O. Connell, J. R. Bliden, K. P. Gandhi, A. Ryan et al., , 2009.

, Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy, JAMA, vol.302, issue.8, pp.849-57

T. Simon, C. Verstuyft, M. Mary-krause, L. Quteineh, E. Drouet et al., Genetic determinants of response to clopidogrel and cardiovascular events, N Engl J Med, vol.360, issue.4, pp.363-75, 2009.
DOI : 10.1056/nejmoa0808227

D. Sibbing, W. Koch, D. Gebhard, T. Schuster, S. Braun et al., Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogrel-treated patients with coronary stent placement, Circulation, vol.121, issue.4, pp.512-520, 2010.
DOI : 10.1161/circulationaha.109.885194

URL : https://www.ahajournals.org/doi/pdf/10.1161/CIRCULATIONAHA.109.885194

L. Bertoletti, E. Ollier, C. Duvillard, X. Delavenne, M. Beyens et al., Direct oral anticoagulants: Current indications and unmet needs in the treatment of venous thromboembolism, Pharmacol Res, vol.118, pp.33-42, 2017.

B. Ingelheim, PRADAXA® : Résumé des caractéristiques du produit, 2017.

. Bayer-healthcare, XARELTO® : Résumé des caractéristiques du produit, 2015.

. Bristol-myers-squibb, ELIQUIS® : Résumé des caractéristiques du produit, 2017.

. Daiichi-sankyo-europe, LIXIANA® : Summary of Product Characteristics

, Information/human/002629/WC500189045.pdf, consulté le 28 août, 2018.

O. Grottke, D. Fries, and B. Nascimento, Perioperatively acquired disorders of coagulation, Curr Opin Anaesthesiol, vol.28, issue.2, pp.113-135, 2015.
DOI : 10.1097/aco.0000000000000176

URL : http://europepmc.org/articles/pmc4418784?pdf=render

H. Autorité-de-santé, LIXIANA® : Avis de la commission de transparence du

, Rapport d'évaluation des médicaments anticoagulants oraux, 2018.

P. Kirchhof, S. Benussi, D. Kotecha, A. Ahlsson, D. Atar et al., 2016 ESC Guidelines for the management of atrial fibrillation developed in collaboration with EACTS, Kardiol Pol, vol.74, issue.12, pp.1359-469, 2016.

G. Cesarman-maus and G. J. Ruiz-argüelles, News in the Indications of Direct Oral Anticoagulants According to the American College of Chest Physicians, Guidelines. Curr Drug Metab, vol.18, issue.7, pp.651-657, 2016.

H. Autorité-de-santé, Prise en charge des surdosages en antivitamines K, des situations à risque hémorragique et des accidents hémorragiques chez les patients traités par antivitamines K en ville et en milieu hospitalier, 2008.

S. Schulman and C. Kearon, Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients, Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis, vol.3, pp.692-696, 2005.

G. F. Tomaselli, K. W. Mahaffey, A. Cuker, P. P. Dobesh, J. U. Doherty et al., , 2017.

, ACC Expert Consensus Decision Pathway on Management of Bleeding in Patients on Oral Anticoagulants: A Report of the American College of Cardiology Task Force on Expert Consensus Decision Pathways, J Am Coll Cardiol, vol.70, issue.24, pp.3042-67

B. T. Samuelson and A. Cuker, Measurement and reversal of the direct oral anticoagulants, Blood Rev, vol.31, issue.1, pp.77-84, 2017.

R. C. Gosselin, D. M. Adcock, S. M. Bates, J. Douxfils, E. J. Favaloro et al., International Council for Standardization in Haematology (ICSH) Recommendations for Laboratory Measurement of Direct Oral Anticoagulants, Thromb Haemost, vol.118, issue.3, pp.437-50, 2018.

R. C. Becker, H. Yang, Y. Barrett, P. Mohan, J. Wang et al., Chromogenic laboratory assays to measure the factor Xa-inhibiting properties of apixaban-an oral, direct and selective factor Xa inhibitor, J Thromb Thrombolysis, vol.32, issue.2, pp.183-190, 2011.

M. M. Samama, G. Contant, T. E. Spiro, E. Perzborn, C. Guinet et al., Evaluation of the anti-factor Xa chromogenic assay for the measurement of rivaroxaban plasma concentrations using calibrators and controls, Thromb Haemost, vol.107, issue.2, pp.379-87, 2012.

P. Albaladejo, F. Bonhomme, N. Blais, J. Collet, D. Faraoni et al., , 2017.

, Management of direct oral anticoagulants in patients undergoing elective surgeries and invasive procedures: Updated guidelines from the French Working Group on Perioperative Hemostasis (GIHP), Anaesth Crit Care Pain Med, vol.36, issue.1, pp.73-79, 2015.

T. E. Warkentin, P. Margetts, S. J. Connolly, A. Lamy, C. Ricci et al., Recombinant factor VIIa (rFVIIa) and hemodialysis to manage massive dabigatranassociated postcardiac surgery bleeding, Blood, vol.119, issue.9, pp.2172-2176, 2012.

P. A. Reilly, T. Lehr, S. Haertter, S. J. Connolly, S. Yusuf et al., The effect of dabigatran plasma concentrations and patient characteristics on the frequency of ischemic stroke and major bleeding in atrial fibrillation patients: the RE-LY Trial (Randomized Evaluation of Long-Term Anticoagulation Therapy), J Am Coll Cardiol, vol.63, issue.4, pp.321-329, 2014.

P. Albaladejo, C. Samama, P. Sié, S. Kauffmann, V. Mémier et al., , 2017.

, Management of Severe Bleeding in Patients Treated with Direct Oral Anticoagulants: An Observational Registry Analysis, Anesthesiology, vol.127, issue.1, pp.111-131

A. Cuker, Laboratory measurement of the non-vitamin K antagonist oral anticoagulants: selecting the optimal assay based on drug, assay availability, and clinical indication, J Thromb Thrombolysis, vol.41, issue.2, pp.241-248, 2016.

T. Cate, H. Henskens, Y. M. Lancé, and M. D. , Practical guidance on the use of laboratory testing in the management of bleeding in patients receiving direct oral anticoagulants, 2017.

. Clinicaltrials, Randomized Evaluation of Long Term Anticoagulant Therapy With Dabigatran Etexilate (RE-LY), 2018.

. Clinicaltrials, An Efficacy and Safety Study of Rivaroxaban With Warfarin for the Prevention of Stroke and Non-Central Nervous System Systemic Embolism in Patients With Non-Valvular Atrial Fibrillation (ROCKET-AF), vol.00403767, 2018.

G. Pernod, P. Albaladejo, A. Godier, C. M. Samama, S. Susen et al., , 2013.

, Management of major bleeding complications and emergency surgery in patients on longterm treatment with direct oral anticoagulants, thrombin or factor-Xa inhibitors: proposals of the working group on perioperative haemostasis (GIHP)-March, Arch Cardiovasc Dis, vol.106, issue.6-7, pp.382-93, 2013.

A. Godier, A. Dincq, A. Martin, A. Radu, I. Leblanc et al., Predictors of pre-procedural concentrations of direct oral anticoagulants: a prospective multicentre study, Eur Heart J, vol.38, issue.31, pp.2431-2440, 2017.

T. Brott, J. Broderick, R. Kothari, W. Barsan, T. Tomsick et al., Early hemorrhage growth in patients with intracerebral hemorrhage, Stroke, vol.28, issue.1, pp.1-5, 1997.

M. A. Miyares, Y. Kuyumjian, S. Eaves, and E. Dollard, Idarucizumab, a Humanized, Monoclonal Antibody Fragment for Immediate Reversal of Dabigatran, J Pharm Pract, vol.28, issue.6, pp.548-54, 2015.

C. V. Pollack, P. A. Reilly, J. Van-ryn, J. W. Eikelboom, S. Glund et al., , 2017.

, Idarucizumab for Dabigatran Reversal-Full Cohort Analysis, N Engl J Med, vol.377, issue.5, pp.431-472

B. Ingelheim, PRAXBIND® : Résumé des caractéristiques du produit, 2016.

S. Glund, V. Moschetti, S. Norris, J. Stangier, M. Schmohl et al., A randomised study in healthy volunteers to investigate the safety, tolerability and pharmacokinetics of idarucizumab, a specific antidote to dabigatran, Thromb Haemost, vol.113, issue.5, pp.943-51, 2015.

W. E. Dager and L. Banares, Reversing the anticoagulation effects of dabigatran, Hosp Pract, vol.45, issue.2, pp.29-38, 2017.
DOI : 10.1080/21548331.2017.1298389

URL : https://touroscholar.touro.edu/author_agreement.pdf

. Clinicaltrials, Reversal of Dabigatran Anticoagulant Effect With Idarucizumab (RE-VERSEAD), 2018.

M. Cappellari, S. Forlivesi, G. M. Squintani, R. Facchinetti, and P. Bovi, Intravenous thrombolysis for stroke after Dabigatran reversal with Idarucizumab: an update, J Thromb Thrombolysis, vol.43, issue.4, pp.528-537, 2017.
DOI : 10.1007/s11239-017-1485-1

F. C. Ng, J. Bice, A. Rodda, M. Lee-archer, and D. E. Crompton, Adverse clinical outcomes after dabigatran reversal with idarucizumab to facilitate acute stroke thrombolysis, J Neurol, vol.264, issue.3, pp.591-595, 2017.
DOI : 10.1007/s00415-017-8410-6

G. Jourdi, I. Gouin-thibault, V. Siguret, S. Gandrille, P. Gaussem et al., FXa-?2Macroglobulin Complex Neutralizes Direct Oral Anticoagulants Targeting FXa In Vitro and In Vivo, Thromb Haemost, vol.118, issue.9, pp.1535-1544, 2018.
DOI : 10.1055/s-0038-1667014

URL : http://www.thieme-connect.de/products/ejournals/pdf/10.1055/s-0038-1667014.pdf

A. Godier, A. Martin, N. Rosencher, and S. Susen, Direct oral anticoagulant associated bleeding, J Mal Vasc, vol.41, issue.4, pp.272-280, 2016.

S. J. Connolly, T. J. Milling, J. W. Eikelboom, C. M. Gibson, J. T. Curnutte et al., , 2016.

, Andexanet Alfa for Acute Major Bleeding Associated with Factor Xa Inhibitors, N Engl J Med, vol.375, issue.12, pp.1131-1172

M. T. Kalathottukaren, A. L. Creagh, S. Abbina, G. Lu, M. J. Karbarz et al., , 2018.

, Comparison of reversal activity and mechanism of action of UHRA, andexanet, and PER977 on heparin and oral FXa inhibitors, Blood Adv, vol.2, issue.16, pp.2104-2118

J. Stangier, K. Rathgen, H. Stähle, and D. Mazur, Influence of renal impairment on the pharmacokinetics and pharmacodynamics of oral dabigatran etexilate: an open-label, parallel-group, single-centre study, Clin Pharmacokinet, vol.49, issue.4, pp.259-68, 2010.

M. Ufer, Comparative efficacy and safety of the novel oral anticoagulants dabigatran, rivaroxaban and apixaban in preclinical and clinical development, Thromb Haemost, vol.103, issue.3, pp.572-85, 2010.

M. Ganetsky, K. M. Babu, S. D. Salhanick, R. S. Brown, and E. W. Boyer, Dabigatran: review of pharmacology and management of bleeding complications of this novel oral anticoagulant, J Med Toxicol, vol.7, issue.4, pp.281-288, 2011.

N. Ishiguro, W. Kishimoto, A. Volz, E. Ludwig-schwellinger, T. Ebner et al., , 2014.

, Impact of endogenous esterase activity on in vitro p-glycoprotein profiling of dabigatran etexilate in Caco-2 monolayers, Drug Metab Dispos, vol.42, issue.2, pp.250-256

S. Blech, T. Ebner, E. Ludwig-schwellinger, J. Stangier, and R. W. , The metabolism and disposition of the oral direct thrombin inhibitor, dabigatran, in humans, Drug Metab Dispos, vol.36, issue.2, pp.386-99, 2008.

O. 'connor, C. T. Kiernan, T. J. Yan, and B. P. , The genetic basis of antiplatelet and anticoagulant therapy: A pharmacogenetic review of newer antiplatelets (clopidogrel, prasugrel and ticagrelor) and anticoagulants (dabigatran, rivaroxaban, apixaban and edoxaban), Expert Opin Drug Metab Toxicol, vol.13, issue.7, pp.725-764, 2017.

S. Harder, Pharmacokinetic and pharmacodynamic evaluation of rivaroxaban: considerations for the treatment of venous thromboembolism, Thromb J, vol.12, p.22, 2014.

P. B. Danielson, The cytochrome P450 superfamily: biochemistry, evolution and drug metabolism in humans, Curr Drug Metab, vol.3, issue.6, pp.561-97, 2002.

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, Pharmacogenomics J, vol.11, issue.4, pp.274-86, 2011.

A. N. Werk and I. Cascorbi, Functional gene variants of CYP3A4, Clin Pharmacol Ther, vol.96, issue.3, pp.340-348, 2014.

. Uniprot, CYP3A4-Cytochrome P450 3A4-Homo sapiens (Human)-CYP3A4 gene & protein, 2018.

. Uniprot, CYP3A5-Cytochrome P450 3A5-Homo sapiens (Human)-CYP3A5 gene & protein, 2018.

H. Hashimoto, K. Toide, R. Kitamura, M. Fujita, S. Tagawa et al., Gene structure of CYP3A4, an adult-specific form of cytochrome P450 in human livers, and its transcriptional control, Eur J Biochem, vol.218, issue.2, pp.585-95, 1993.

, NCBI. rs35599367 : Reference SNP Cluster Report, 2018.

P. Kuehl, J. Zhang, Y. Lin, J. Lamba, M. Assem et al., Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression, Nat Genet, vol.27, issue.4, pp.383-91, 2001.

N. , Reference SNP Cluster Report ** With drug-response allele **, 2018.

H. Fukushima-uesaka, Y. Saito, H. Watanabe, K. Shiseki, M. Saeki et al., , 2004.

, Haplotypes of CYP3A4 and their close linkage with CYP3A5 haplotypes in a Japanese population, Hum Mutat, vol.23, issue.1, p.100

T. S. Klose, J. A. Blaisdell, and J. A. Goldstein, Gene structure of CYP2C8 and extrahepatic distribution of the human CYP2Cs, J Biochem Mol Toxicol, vol.13, issue.6, pp.289-95, 1999.

Y. Chen and J. A. Goldstein, The transcriptional regulation of the human CYP2C genes, Curr Drug Metab, vol.10, issue.6, pp.567-78, 2009.

J. Mwinyi, I. Cavaco, R. S. Pedersen, A. Persson, S. Burkhardt et al., Regulation of CYP2C19 expression by estrogen receptor ?: implications for estrogendependent inhibition of drug metabolism, Mol Pharmacol, vol.78, issue.5, pp.886-94, 2010.

. Uniprot, CYP2C19-Cytochrome P450 2C19-Homo sapiens (Human)-CYP2C19 gene & protein, 2018.

S. M. De-morais, G. R. Wilkinson, J. Blaisdell, K. Nakamura, U. A. Meyer et al., The major genetic defect responsible for the polymorphism of S-mephenytoin metabolism in humans, J Biol Chem, vol.269, issue.22, pp.15419-15441, 1994.

S. A. Scott, K. Sangkuhl, A. R. Shuldiner, J. Hulot, C. F. Thorn et al., PharmGKB summary: very important pharmacogene information for cytochrome P450, family 2, subfamily C, polypeptide 19, Pharmacogenet Genomics, vol.22, issue.2, pp.159-65, 2012.

M. Bodor, E. J. Kelly, and R. J. Ho, Characterization of the human MDR1 gene, AAPS J, vol.7, issue.1, pp.1-5, 2005.

U. Brinkmann and M. Eichelbaum, Polymorphisms in the ABC drug transporter gene MDR1, Pharmacogenomics J, vol.1, issue.1, pp.59-64, 2001.

H. T. Idriss, Y. A. Hannun, E. Boulpaep, and S. Basavappa, Regulation of volume-activated chloride channels by P-glycoprotein: phosphorylation has the final say, J Physiol, vol.524, issue.3, pp.629-665, 2000.

S. G. Aller, J. Yu, A. Ward, Y. Weng, S. Chittaboina et al., Structure of Pglycoprotein reveals a molecular basis for poly-specific drug binding, Science, vol.323, issue.5922, pp.1718-1740, 2009.

. Uniprot, ABCB1-Multidrug resistance protein 1-Homo sapiens (Human)-ABCB1 gene & protein, 2018.

L. M. Hodges, S. M. Markova, L. W. Chinn, J. M. Gow, D. L. Kroetz et al., Very important pharmacogene summary: ABCB1 (MDR1, P-glycoprotein), Pharmacogenet Genomics, vol.21, issue.3, pp.152-61, 2011.

G. D. Leschziner, T. Andrew, M. Pirmohamed, and M. R. Johnson, ABCB1 genotype and PGP expression, function and therapeutic drug response: a critical review and recommendations for future research, Pharmacogenomics J, vol.7, issue.3, pp.154-79, 2007.
DOI : 10.1038/sj.tpj.6500413

URL : https://www.nature.com/articles/6500413.pdf

, NCBI. rs1128503 : Reference SNP Cluster Report ** With Benign allele **, 2018.

, NCBI. rs2032582 : Reference SNP Cluster Report ** With drug-response, 2018.

, NCBI. rs1045642 : Reference SNP Cluster Report ** With drug-response allele **, 2018.

, NCBI. rs4148738 : Reference SNP Cluster Report, 2018.

R. B. Kim, B. F. Leake, E. F. Choo, G. K. Dresser, S. V. Kubba et al., Identification of functionally variant MDR1 alleles among European Americans and African Americans, 2001.

, Clin Pharmacol Ther, vol.70, issue.2, pp.189-99

D. L. Kroetz, C. Pauli-magnus, L. M. Hodges, C. C. Huang, M. Kawamoto et al., Sequence diversity and haplotype structure in the human ABCB1 (MDR1, multidrug resistance transporter) gene, Pharmacogenetics, vol.13, issue.8, pp.481-94, 2003.

D. Wang, A. D. Johnson, A. C. Papp, D. L. Kroetz, and W. Sadée, Multidrug resistance polypeptide 1 (MDR1, ABCB1) variant 3435C>T affects mRNA stability, Pharmacogenet Genomics, vol.15, issue.10, pp.693-704, 2005.

I. Gouin-thibault, X. Delavenne, A. Blanchard, V. Siguret, J. E. Salem et al., , 2017.

, Interindividual variability in dabigatran and rivaroxaban exposure: contribution of ABCB1 genetic polymorphisms and interaction with clarithromycin, J Thromb Haemost, vol.15, issue.2, pp.273-83

L. Wu, H. Zhao, C. Wen, Y. Li, Y. Shao et al., Combined Influence of Genetic Polymorphism and DNA Methylation on ABCB1 Expression and Function in Healthy Chinese Males, Eur J Drug Metab Pharmacokinet, vol.42, issue.4, pp.627-634, 2016.

Z. Merali, R. S. Paré, and G. , The pharmacogenetics of carboxylesterases: CES1 and CES2 genetic variants and their clinical effect, Drug Metabol Drug Interact, vol.29, issue.3, pp.143-51, 2014.

J. Shi, X. Wang, J. Nguyen, B. E. Bleske, Y. Liang et al., Dabigatran etexilate activation is affected by the CES1 genetic polymorphism G143E (rs71647871) and gender, Biochem Pharmacol, vol.119, pp.76-84, 2016.

G. Paré, N. Eriksson, T. Lehr, S. Connolly, J. Eikelboom et al., Genetic determinants of dabigatran plasma levels and their relation to bleeding, Circulation, vol.127, issue.13, pp.1404-1416, 2013.

. Uniprot, CES1-Liver carboxylesterase 1 precursor-Homo sapiens (Human)-CES1 gene & protein, 2018.

D. A. Sychev, A. N. Levanov, T. V. Shelekhova, P. O. Bochkov, N. P. Denisenko et al., The impact of ABCB1 (rs1045642 and rs4148738) and CES1 (rs2244613) gene polymorphisms on dabigatran equilibrium peak concentration in patients after total knee arthroplasty, Pharmgenomics Pers Med, vol.11, pp.127-164, 2018.

C. Dimatteo, D. 'andrea, G. Vecchione, G. Paoletti, O. Cappucci et al., , 2016.

, Pharmacogenetics of dabigatran etexilate interindividual variability, Thromb Res, vol.144, pp.1-5

K. Ing-lorenzini, Y. Daali, P. Fontana, J. Desmeules, and C. Samer, Rivaroxaban-Induced Hemorrhage Associated with ABCB1 Genetic Defect, Front Pharmacol, vol.7, p.494, 2016.

D. A. Sychev, A. Vardanyan, A. Rozhkov, E. Hachatryan, A. Badanyan et al., , 2018.

, CYP3A Activity and Rivaroxaban Serum Concentrations in Russian Patients with Deep Vein Thrombosis, Genet Test Mol Biomarkers, vol.22, issue.1, pp.51-55

D. Dai, J. Tang, R. Rose, E. Hodgson, R. J. Bienstock et al., , 2001.

, Identification of variants of CYP3A4 and characterization of their abilities to metabolize testosterone and chlorpyrifos, J Pharmacol Exp Ther, vol.299, issue.3, pp.825-856

C. Dimatteo, D. 'andrea, G. Vecchione, G. Paoletti, O. Tiscia et al., , 2016.

, ABCB1 SNP rs4148738 modulation of apixaban interindividual variability, Thromb Res, vol.145, pp.24-30

S. Ueshima, D. Hira, R. Fujii, Y. Kimura, C. Tomitsuka et al., Impact of ABCB1, ABCG2, and CYP3A5 polymorphisms on plasma trough concentrations of apixaban in Japanese patients with atrial fibrillation, Pharmacogenet Genomics, vol.27, issue.9, pp.329-365, 2017.

A. V. Kryukov, D. A. Sychev, D. A. Andreev, K. A. Ryzhikova, E. A. Grishina et al., Influence of ABCB1 and CYP3A5 gene polymorphisms on pharmacokinetics of apixaban in patients with atrial fibrillation and acute stroke, Pharmgenomics Pers Med, vol.11, pp.43-52, 2018.

A. A?i?, D. Marjanovi?, J. Mirat, and D. Primorac, Pharmacogenetics of novel oral anticoagulants: a review of identified gene variants & future perspectives, Per Med, vol.15, issue.3, pp.209-230, 2018.

A. G. Vandell, J. Lee, M. Shi, I. Rubets, K. S. Brown et al., An integrated pharmacokinetic/pharmacogenomic analysis of ABCB1 and SLCO1B1 polymorphisms on edoxaban exposure, Pharmacogenomics J, vol.18, issue.1, pp.153-162, 2018.

A. Sennesael, N. Panin, C. Vancraeynest, L. Pochet, A. Spinewine et al., Effect of ABCB1 genetic polymorphisms on the transport of rivaroxaban in HEK293 recombinant cell lines, Sci Rep, vol.8, issue.1, p.10514, 2018.

E. Gautier-veyret, X. Fonrose, J. Tonini, A. Thiebaut-bertrand, M. Bartoli et al., Variability of voriconazole plasma concentrations after allogeneic hematopoietic stem cell transplantation: impact of cytochrome p450 polymorphisms and comedications on initial and subsequent trough levels, Antimicrob Agents Chemother, vol.59, issue.4, pp.2305-2319, 2015.

H. Autorité-de-santé, Transfusions de globules rouges homologues : produits, indications, alternatives, 2014.

F. Lamoureux, T. Duflot, J. Woillard, D. Metsu, T. Pereira et al., Impact of CYP2C19 genetic polymorphisms on voriconazole dosing and exposure in adult patients with invasive fungal infections, Int J Antimicrob Agents, vol.47, issue.2, pp.124-155, 2016.

T. Duflot, A. Schrapp, J. Bellien, and F. Lamoureux, Impact of CYP3A4 Genotype on Voriconazole Exposure, Clin Pharmacol Ther, vol.103, issue.2, pp.185-191, 2018.

S. Ueshima, D. Hira, Y. Kimura, R. Fujii, C. Tomitsuka et al., Population pharmacokinetics and pharmacogenomics of apixaban in Japanese adult patients with atrial fibrillation, Br J Clin Pharmacol, vol.84, issue.6, pp.1301-1313, 2018.

M. Whirl-carrillo, E. M. Mcdonagh, J. M. Hebert, L. Gong, K. Sangkuhl et al., Pharmacogenomics knowledge for personalized medicine, Clin Pharmacol Ther, vol.92, issue.4, pp.414-421, 2012.

P. Derogis, L. R. Sanches, V. F. De-aranda, M. P. Colombini, C. Mangueira et al., Determination of rivaroxaban in patient's plasma samples by anti-Xa chromogenic test associated to High Performance Liquid Chromatography tandem Mass Spectrometry (HPLC-MS/MS), PLoS ONE, vol.12, issue.2, p.171272, 2017.

M. Skeppholm, F. Al-aieshy, M. Berndtsson, F. Al-khalili, Y. Rönquist-nii et al., Clinical evaluation of laboratory methods to monitor apixaban treatment in patients with atrial fibrillation, Thromb Res, vol.136, issue.1, pp.148-53, 2015.

J. Douxfils, A. Tamigniau, B. Chatelain, C. Chatelain, P. Wallemacq et al., Comparison of calibrated chromogenic anti-Xa assay and PT tests with LC-MS/MS for the therapeutic monitoring of patients treated with rivaroxaban, Thromb Haemost, vol.110, issue.4, pp.723-754, 2013.

, Evaluation of glomerular filtration rate and proteinuria for the diagnosis of chronic kidney disease, Nephrol Ther, vol.5, issue.4, pp.302-307, 2009.

N. Sablani, J. Garg, B. Hasan, R. Patel, and M. W. Martinez, First reported case series in the United States of hemopericardium in patients on apixaban, HeartRhythm Case Rep, vol.4, issue.2, pp.82-86, 2017.

L. Guize, F. Thomas, K. Bean, A. Benetos, and B. Pannier, Atrial fibrillation: prevalence, risk factors and mortality in a large French population with 15 years of follow-up, Bull Acad Natl Med, vol.191, issue.4-5, pp.791-805, 2007.

N. De-crem, K. Peerlinck, T. Vanassche, K. Vanheule, B. Debaveye et al., , 2015.

, Abnormal uterine bleeding in VTE patients treated with rivaroxaban compared to vitamin K antagonists, Thromb Res, vol.136, issue.4, pp.749-53

M. Ferreira, S. Barsam, J. P. Patel, J. Czuprynska, L. N. Roberts et al., Heavy menstrual bleeding on rivaroxaban, Br J Haematol, vol.173, issue.2, pp.314-319, 2016.

B. Myers and A. Webster, Heavy menstrual bleeding on Rivaroxaban-Comparison with Apixaban, Br J Haematol, vol.176, issue.5, pp.833-838, 2017.

S. Eisho, N. M. Salem, J. L. Hoffman, J. M. Koerber, and M. A. Smythe, Major bleeding with apixaban in atrial fibrillation: patient characteristics, management, and outcomes, Hosp Pract, pp.1-5, 1995.

S. Frol, P. Oblak, and J. , Early Outcome after Intracranial Hemorrhage Related to Non-Vitamin K Oral Anticoagulants, Interv Neurol, vol.7, issue.1-2, pp.19-25, 2018.

L. H. Cavallari, J. Shin, and M. A. Perera, Role of pharmacogenomics in the management of traditional and novel oral anticoagulants, Pharmacotherapy, issue.12, pp.1192-207, 2011.

A. I. Mazur-bialy, K. Zdebska, E. Wypasek, and A. Undas, Repeated bleeding complications during therapy with vitamin K antagonists in a patient with the VKORC1*2A and the CYP2C9*3/*3 alleles: genetic testing to support switching to new oral anticoagulants, Thromb Res, vol.131, issue.3, pp.279-80, 2013.

N. Cullell, C. Carrera, E. Muiño, N. Torres, J. Krupinski et al., , 2018.

, Pharmacogenetic studies with oral anticoagulants. Genome-wide association studies in vitamin K antagonist and direct oral anticoagulants, Oncotarget, vol.9, issue.49, pp.29238-58