메뉴 건너뛰기




Volumn 100, Issue 3, 2016, Pages 295-304

Coadministration of ticagrelor and ritonavir: Toward prospective dose adjustment to maintain an optimal platelet inhibition using the PBPK approach

Author keywords

[No Author keywords available]

Indexed keywords

DRUG METABOLITE; KETOCONAZOLE; RITONAVIR; TICAGRELOR; ADENOSINE; ANTITHROMBOCYTIC AGENT; CYTOCHROME P450 3A INHIBITOR; HUMAN IMMUNODEFICIENCY VIRUS PROTEINASE INHIBITOR;

EID: 84988925087     PISSN: 00099236     EISSN: 15326535     Source Type: Journal    
DOI: 10.1002/cpt.407     Document Type: Article
Times cited : (38)

References (50)
  • 1
    • 78650289551 scopus 로고    scopus 로고
    • Adenosine release: a potential explanation for the benefits of ticagrelor in the PLATelet inhibition and clinical outcomes trial?
    • Serebruany, V.L. Adenosine release: a potential explanation for the benefits of ticagrelor in the PLATelet inhibition and clinical outcomes trial? Am. Heart J. 161, 1–4 (2011).
    • (2011) Am. Heart J. , vol.161 , pp. 1-4
    • Serebruany, V.L.1
  • 2
    • 0347416933 scopus 로고    scopus 로고
    • Acute myocardial infarction in patients infected with human immunodeficiency virus
    • Varriale, P., Saravi, G., Hernandez, E. & Carbon, F. Acute myocardial infarction in patients infected with human immunodeficiency virus. Am. Heart J. 147, 55–59 (2004).
    • (2004) Am. Heart J. , vol.147 , pp. 55-59
    • Varriale, P.1    Saravi, G.2    Hernandez, E.3    Carbon, F.4
  • 3
    • 0038466343 scopus 로고    scopus 로고
    • Coronary artery disease in HIV infected patients
    • Escaut, L. et al. Coronary artery disease in HIV infected patients. Intensive Care Med. 29, 969–973 (2003).
    • (2003) Intensive Care Med. , vol.29 , pp. 969-973
    • Escaut, L.1
  • 4
    • 34250671221 scopus 로고    scopus 로고
    • Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease
    • Triant, V.A., Lee, H., Hadigan, C. & Grinspoon, S.K. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J. Clin. Endocrinol. Metab. 92, 2506–2512 (2007).
    • (2007) J. Clin. Endocrinol. Metab. , vol.92 , pp. 2506-2512
    • Triant, V.A.1    Lee, H.2    Hadigan, C.3    Grinspoon, S.K.4
  • 7
    • 84876713253 scopus 로고    scopus 로고
    • Effect of rifampicin on the pharmacokinetics and pharmacodynamics of ticagrelor in healthy subjects
    • Teng, R., Mitchell, P. & Butler, K. Effect of rifampicin on the pharmacokinetics and pharmacodynamics of ticagrelor in healthy subjects. Eur. J. Clin. Pharmacol. 69, 877–883 (2013).
    • (2013) Eur. J. Clin. Pharmacol. , vol.69 , pp. 877-883
    • Teng, R.1    Mitchell, P.2    Butler, K.3
  • 8
    • 84971663950 scopus 로고    scopus 로고
    • Morphine delays and attenuates ticagrelor exposure and action in patients with myocardial infarction: the randomized, double-blind, placebo-controlled IMPRESSION trial
    • Kubica, J. et al. Morphine delays and attenuates ticagrelor exposure and action in patients with myocardial infarction: the randomized, double-blind, placebo-controlled IMPRESSION trial. Eur. Heart J. 37, 245–252 (2016).
    • (2016) Eur. Heart J. , vol.37 , pp. 245-252
    • Kubica, J.1
  • 9
    • 84878117557 scopus 로고    scopus 로고
    • Grapefruit juice markedly increases the plasma concentrations and antiplatelet effects of ticagrelor in healthy subjects
    • Holmberg, M.T. et al. Grapefruit juice markedly increases the plasma concentrations and antiplatelet effects of ticagrelor in healthy subjects. Br. J. Clin. Pharmacol. 75, 1488–1496 (2013).
    • (2013) Br. J. Clin. Pharmacol. , vol.75 , pp. 1488-1496
    • Holmberg, M.T.1
  • 11
    • 77955992168 scopus 로고    scopus 로고
    • Absorption, distribution, metabolism, and excretion of ticagrelor in healthy subjects
    • Teng, R., Oliver, S., Hayes, M.A. & Butler, K. Absorption, distribution, metabolism, and excretion of ticagrelor in healthy subjects. Drug Metab. Dispos. 38, 1514–1521 (2010).
    • (2010) Drug Metab. Dispos. , vol.38 , pp. 1514-1521
    • Teng, R.1    Oliver, S.2    Hayes, M.A.3    Butler, K.4
  • 12
    • 84928887973 scopus 로고    scopus 로고
    • Predicting the effect of cytochrome P450 inhibitors on substrate drugs: analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration
    • Wagner, C. et al. Predicting the effect of cytochrome P450 inhibitors on substrate drugs: analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration. Clin. Pharmacokinet. 54, 117–127 (2015).
    • (2015) Clin. Pharmacokinet. , vol.54 , pp. 117-127
    • Wagner, C.1
  • 13
    • 77954468084 scopus 로고    scopus 로고
    • Pharmacokinetics, pharmacodynamics, tolerability and safety of single ascending doses of ticagrelor, a reversibly binding oral P2Y(12) receptor antagonist, in healthy subjects
    • Teng, R. & Butler, K. Pharmacokinetics, pharmacodynamics, tolerability and safety of single ascending doses of ticagrelor, a reversibly binding oral P2Y(12) receptor antagonist, in healthy subjects. Eur. J. Clin. Pharmacol. 66, 487–496 (2010).
    • (2010) Eur. J. Clin. Pharmacol. , vol.66 , pp. 487-496
    • Teng, R.1    Butler, K.2
  • 14
    • 84885062087 scopus 로고    scopus 로고
    • Safety, tolerability, pharmacokinetics and pharmacodynamics of high single-ascending doses of ticagrelor in healthy volunteers
    • Teng, R. & Butler, K. Safety, tolerability, pharmacokinetics and pharmacodynamics of high single-ascending doses of ticagrelor in healthy volunteers. Int. J. Clin. Pharmacol. Ther. 51, 795–806 (2013).
    • (2013) Int. J. Clin. Pharmacol. Ther. , vol.51 , pp. 795-806
    • Teng, R.1    Butler, K.2
  • 15
    • 84890025521 scopus 로고    scopus 로고
    • Consensus and update on the definition of on-treatment platelet reactivity to adenosine diphosphate associated with ischemia and bleeding
    • Tantry, U.S. et al. Consensus and update on the definition of on-treatment platelet reactivity to adenosine diphosphate associated with ischemia and bleeding. J. Am. Coll. Cardiol. 62, 2261–2273 (2013).
    • (2013) J. Am. Coll. Cardiol. , vol.62 , pp. 2261-2273
    • Tantry, U.S.1
  • 16
    • 84927172074 scopus 로고    scopus 로고
    • Predicting drug-drug interactions involving multiple mechanisms using physiologically based pharmacokinetic modeling: a case study with ruxolitinib
    • Shi, J.G., Fraczkiewicz, G., Williams, W.V. & Yeleswaram, S. Predicting drug-drug interactions involving multiple mechanisms using physiologically based pharmacokinetic modeling: a case study with ruxolitinib. Clin. Pharmacol. Ther. 97, 177–185 (2015).
    • (2015) Clin. Pharmacol. Ther. , vol.97 , pp. 177-185
    • Shi, J.G.1    Fraczkiewicz, G.2    Williams, W.V.3    Yeleswaram, S.4
  • 17
    • 84959081464 scopus 로고    scopus 로고
    • Physiologically-based pharmacokinetic modeling of macitentan: prediction of drug-drug interactions
    • de Kanter, R. et al. Physiologically-based pharmacokinetic modeling of macitentan: prediction of drug-drug interactions. Clin. Pharmacokinet. 55, 369–380 (2016).
    • (2016) Clin. Pharmacokinet. , vol.55 , pp. 369-380
    • de Kanter, R.1
  • 18
    • 84891059562 scopus 로고    scopus 로고
    • Effect of the CYP3A inhibitors, diltiazem and ketoconazole, on ticagrelor pharmacokinetics in healthy volunteers
    • Teng, R. & Butler, K. Effect of the CYP3A inhibitors, diltiazem and ketoconazole, on ticagrelor pharmacokinetics in healthy volunteers. J. Drug Assessment 2, 30–39 (2013).
    • (2013) J. Drug Assessment , vol.2 , pp. 30-39
    • Teng, R.1    Butler, K.2
  • 20
    • 77953498801 scopus 로고    scopus 로고
    • Pharmacokinetics, pharmacodynamics, safety and tolerability of multiple ascending doses of ticagrelor in healthy volunteers
    • Butler, K. & Teng, R. Pharmacokinetics, pharmacodynamics, safety and tolerability of multiple ascending doses of ticagrelor in healthy volunteers. Br. J. Clin. Pharmacol. 70, 65–77 (2010).
    • (2010) Br. J. Clin. Pharmacol. , vol.70 , pp. 65-77
    • Butler, K.1    Teng, R.2
  • 21
    • 80053530180 scopus 로고    scopus 로고
    • Vectorial transport of fexofenadine across Caco-2 cells: involvement of apical uptake and basolateral efflux transporters
    • Ming, X., Knight, B.M. & Thakker D.R. Vectorial transport of fexofenadine across Caco-2 cells: involvement of apical uptake and basolateral efflux transporters. Mol. Pharm. 8, 1677–1686 (2011).
    • (2011) Mol. Pharm. , vol.8 , pp. 1677-1686
    • Ming, X.1    Knight, B.M.2    Thakker, D.R.3
  • 22
    • 24944482460 scopus 로고    scopus 로고
    • Contribution of OATP (organic anion-transporting polypeptide) family transporters to the hepatic uptake of fexofenadine in humans
    • Shimizu, M. et al. Contribution of OATP (organic anion-transporting polypeptide) family transporters to the hepatic uptake of fexofenadine in humans. Drug Metab. Dispos. 33, 1477–1481 (2005).
    • (2005) Drug Metab. Dispos. , vol.33 , pp. 1477-1481
    • Shimizu, M.1
  • 23
    • 11344279702 scopus 로고    scopus 로고
    • Different effects of three transporting inhibitors, verapamil, cimetidine, and probenecid, on fexofenadine pharmacokinetics
    • Yasui-Furukori, N., Uno, T., Sugawara, K. & Tateishi, T. Different effects of three transporting inhibitors, verapamil, cimetidine, and probenecid, on fexofenadine pharmacokinetics. Clin. Pharmacol. Ther. 77, 17–23 (2005).
    • (2005) Clin. Pharmacol. Ther. , vol.77 , pp. 17-23
    • Yasui-Furukori, N.1    Uno, T.2    Sugawara, K.3    Tateishi, T.4
  • 24
    • 84880271700 scopus 로고    scopus 로고
    • Mechanisms of pharmacokinetic enhancement between ritonavir and saquinavir; micro/small dosing tests using midazolam (CYP3A4), fexofenadine (p-glycoprotein), and pravastatin (OATP1B1) as probe drugs
    • Ieiri, I. et al. Mechanisms of pharmacokinetic enhancement between ritonavir and saquinavir; micro/small dosing tests using midazolam (CYP3A4), fexofenadine (p-glycoprotein), and pravastatin (OATP1B1) as probe drugs. J. Clin. Pharmacol. 53, 654–661 (2013).
    • (2013) J. Clin. Pharmacol. , vol.53 , pp. 654-661
    • Ieiri, I.1
  • 25
    • 77956247876 scopus 로고    scopus 로고
    • pH dependence of organic anion-transporting polypeptide 2B1 in Caco-2 cells: potential role in antiretroviral drug oral bioavailability and drug-drug interactions
    • Kis, O., Zastre, J.A., Ramaswamy, M. & Bendayan, R. pH dependence of organic anion-transporting polypeptide 2B1 in Caco-2 cells: potential role in antiretroviral drug oral bioavailability and drug-drug interactions. J. Pharmacol. Exp. Ther. 334, 1009–1022 (2010).
    • (2010) J. Pharmacol. Exp. Ther. , vol.334 , pp. 1009-1022
    • Kis, O.1    Zastre, J.A.2    Ramaswamy, M.3    Bendayan, R.4
  • 26
    • 77149153424 scopus 로고    scopus 로고
    • Interaction of HIV protease inhibitors with OATP1B1, 1B3, and 2B1
    • Annaert, P., Ye, Z.W., Stieger, B. & Augustijns P. Interaction of HIV protease inhibitors with OATP1B1, 1B3, and 2B1. Xenobiotica 40, 163–176 (2010).
    • (2010) Xenobiotica , vol.40 , pp. 163-176
    • Annaert, P.1    Ye, Z.W.2    Stieger, B.3    Augustijns, P.4
  • 27
    • 78651069086 scopus 로고    scopus 로고
    • Do activities of cytochrome P450 (CYP)3A, CYP2D6 and P-glycoprotein differ between healthy volunteers and HIV-infected patients?
    • Jetter, A. et al. Do activities of cytochrome P450 (CYP)3A, CYP2D6 and P-glycoprotein differ between healthy volunteers and HIV-infected patients? Antivir. Ther. 15, 975–983 (2010).
    • (2010) Antivir. Ther. , vol.15 , pp. 975-983
    • Jetter, A.1
  • 28
    • 77954461644 scopus 로고    scopus 로고
    • Variability in drug metabolizing enzyme activity in HIV-infected patients
    • Jones, A.E. et al. Variability in drug metabolizing enzyme activity in HIV-infected patients. Eur. J. Clin. Pharmacol. 66, 475–485 (2010).
    • (2010) Eur. J. Clin. Pharmacol. , vol.66 , pp. 475-485
    • Jones, A.E.1
  • 29
    • 84873581378 scopus 로고    scopus 로고
    • Potent P2Y12 receptor inhibitors in patients with acute coronary syndrome. Agents, indications, issues to consider in clinical practice
    • Sibbing, D., Orban, M. & Massberg S. Potent P2Y12 receptor inhibitors in patients with acute coronary syndrome. Agents, indications, issues to consider in clinical practice. Hamostaseologie 33, 9–15 (2013).
    • (2013) Hamostaseologie , vol.33 , pp. 9-15
    • Sibbing, D.1    Orban, M.2    Massberg, S.3
  • 30
    • 84900403669 scopus 로고    scopus 로고
    • A patient with HIV and tuberculosis with diminished clopidogrel response
    • Metzger, N.L. & Momary K.M. A patient with HIV and tuberculosis with diminished clopidogrel response. Int. J. STD AIDS 25, 532–534 (2014).
    • (2014) Int. J. STD AIDS , vol.25 , pp. 532-534
    • Metzger, N.L.1    Momary, K.M.2
  • 31
    • 80054966619 scopus 로고    scopus 로고
    • Ritonavir inhibits the two main prasugrel bioactivation pathways in vitro: a potential drug-drug interaction in HIV patients
    • Daali, Y., Ancrenaz, V., Bosilkovska, M., Dayer, P. & Desmeules, J. Ritonavir inhibits the two main prasugrel bioactivation pathways in vitro: a potential drug-drug interaction in HIV patients. Metabolism 60, 1584–1589 (2011).
    • (2011) Metabolism , vol.60 , pp. 1584-1589
    • Daali, Y.1    Ancrenaz, V.2    Bosilkovska, M.3    Dayer, P.4    Desmeules, J.5
  • 32
    • 84872151682 scopus 로고    scopus 로고
    • Pharmacokinetic interaction between prasugrel and ritonavir in healthy volunteers
    • Ancrenaz, V. et al. Pharmacokinetic interaction between prasugrel and ritonavir in healthy volunteers. Basic Clin. Pharmacol. Toxicol. 112, 132–137 (2013).
    • (2013) Basic Clin. Pharmacol. Toxicol. , vol.112 , pp. 132-137
    • Ancrenaz, V.1
  • 33
    • 79952831685 scopus 로고    scopus 로고
    • In vitro evaluation of potential drug-drug interactions with ticagrelor: cytochrome P450 reaction phenotyping, inhibition, induction, and differential kinetics
    • Zhou, D., Andersson, T.B. & Grimm S.W. In vitro evaluation of potential drug-drug interactions with ticagrelor: cytochrome P450 reaction phenotyping, inhibition, induction, and differential kinetics. Drug Metab. Dispos. 39, 703–710 (2011).
    • (2011) Drug Metab. Dispos. , vol.39 , pp. 703-710
    • Zhou, D.1    Andersson, T.B.2    Grimm, S.W.3
  • 34
    • 67649392513 scopus 로고    scopus 로고
    • A framework for assessing inter-individual variability in pharmacokinetics using virtual human populations and integrating general knowledge of physical chemistry, biology, anatomy, physiology and genetics: a tale of ‘bottom-up’ vs ‘top-down’ recognition of covariates
    • Jamei, M., Dickinson, G.L. & Rostami-Hodjegan, A. A framework for assessing inter-individual variability in pharmacokinetics using virtual human populations and integrating general knowledge of physical chemistry, biology, anatomy, physiology and genetics: a tale of ‘bottom-up’ vs ‘top-down’ recognition of covariates. Drug Metab. Pharmacokinet. 24, 53–75 (2009).
    • (2009) Drug Metab. Pharmacokinet. , vol.24 , pp. 53-75
    • Jamei, M.1    Dickinson, G.L.2    Rostami-Hodjegan, A.3
  • 35
    • 33947587274 scopus 로고    scopus 로고
    • Simulation and prediction of in vivo drug metabolism in human populations from in vitro data
    • Rostami-Hodjegan, A. & Tucker G.T. Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat. Rev. Drug Discov. 6, 140–148 (2007).
    • (2007) Nat. Rev. Drug Discov. , vol.6 , pp. 140-148
    • Rostami-Hodjegan, A.1    Tucker, G.T.2
  • 36
    • 13244287685 scopus 로고    scopus 로고
    • ‘In silico’ simulations to assess the ‘in vivo’ consequences of ‘in vitro’ metabolic drug-drug interactions
    • Rostami-Hodjegan, A. & Tucker G.T. ‘In silico’ simulations to assess the ‘in vivo’ consequences of ‘in vitro’ metabolic drug-drug interactions. Drug Discov. Today Technol. 1, 441–448 (2004).
    • (2004) Drug Discov. Today Technol. , vol.1 , pp. 441-448
    • Rostami-Hodjegan, A.1    Tucker, G.T.2
  • 37
    • 4344576481 scopus 로고    scopus 로고
    • A simplified analytical method for a phenotyping cocktail of major CYP450 biotransformation routes
    • Jerdi, M.C., Daali, Y., Oestreicher, M.K., Cherkaoui, S. & Dayer, P. A simplified analytical method for a phenotyping cocktail of major CYP450 biotransformation routes. J. Pharm. Biomed. Anal. 35, 1203–1212 (2004).
    • (2004) J. Pharm. Biomed. Anal. , vol.35 , pp. 1203-1212
    • Jerdi, M.C.1    Daali, Y.2    Oestreicher, M.K.3    Cherkaoui, S.4    Dayer, P.5
  • 38
    • 84892698531 scopus 로고    scopus 로고
    • Simultaneous LC-MS/MS quantification of P-glycoprotein and cytochrome P450 probe substrates and their metabolites in DBS and plasma
    • Bosilkovska, M. et al. Simultaneous LC-MS/MS quantification of P-glycoprotein and cytochrome P450 probe substrates and their metabolites in DBS and plasma. Bioanalysis 6, 151–164 (2014).
    • (2014) Bioanalysis , vol.6 , pp. 151-164
    • Bosilkovska, M.1
  • 40
    • 73949112820 scopus 로고    scopus 로고
    • Randomized double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary artery disease: the ONSET/OFFSET study
    • Gurbel, P.A. et al. Randomized double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary artery disease: the ONSET/OFFSET study. Circulation 120, 2577–2585 (2009).
    • (2009) Circulation , vol.120 , pp. 2577-2585
    • Gurbel, P.A.1
  • 41
    • 77249098703 scopus 로고    scopus 로고
    • Clopidogrel response: head-to-head comparison of different platelet assays to identify clopidogrel non responder patients after coronary stenting
    • Cuisset, T. et al. Clopidogrel response: head-to-head comparison of different platelet assays to identify clopidogrel non responder patients after coronary stenting. Arch. Cardiovasc. Dis. 103, 39–45 (2010).
    • (2010) Arch. Cardiovasc. Dis. , vol.103 , pp. 39-45
    • Cuisset, T.1
  • 42
    • 66149114859 scopus 로고    scopus 로고
    • Comparison of VerifyNow-P2Y12 test and Flow Cytometry for monitoring individual platelet response to clopidogrel. What is the cut-off value for identifying patients who are low responders to clopidogrel therapy?
    • Godino, C. et al. Comparison of VerifyNow-P2Y12 test and Flow Cytometry for monitoring individual platelet response to clopidogrel. What is the cut-off value for identifying patients who are low responders to clopidogrel therapy? Thromb. J. 7, 4 (2009).
    • (2009) Thromb. J. , vol.7 , pp. 4
    • Godino, C.1
  • 43
    • 77149128031 scopus 로고    scopus 로고
    • VerifyNow and VASP phosphorylation assays give similar results for patients receiving clopidogrel, but they do not always correlate with platelet aggregation
    • Bidet, A. et al. VerifyNow and VASP phosphorylation assays give similar results for patients receiving clopidogrel, but they do not always correlate with platelet aggregation. Platelets 21, 94–100 (2010).
    • (2010) Platelets , vol.21 , pp. 94-100
    • Bidet, A.1
  • 44
    • 33845366186 scopus 로고    scopus 로고
    • Comparison of VASP-phosphorylation assay to light-transmission aggregometry in assessing inhibition of the platelet ADP P2Y12 receptor
    • Pampuch, A., Cerletti, C. & de Gaetano, G. Comparison of VASP-phosphorylation assay to light-transmission aggregometry in assessing inhibition of the platelet ADP P2Y12 receptor. Thromb. Haemost. 96, 767–773 (2006).
    • (2006) Thromb. Haemost. , vol.96 , pp. 767-773
    • Pampuch, A.1    Cerletti, C.2    de Gaetano, G.3
  • 45
    • 38349168418 scopus 로고    scopus 로고
    • A comparison of the antiplatelet effects of prasugrel and high-dose clopidogrel as assessed by VASP-phosphorylation and light transmission aggregometry
    • Jakubowski, J.A. et al. A comparison of the antiplatelet effects of prasugrel and high-dose clopidogrel as assessed by VASP-phosphorylation and light transmission aggregometry. Thromb. Haemost. 99, 215–222 (2008).
    • (2008) Thromb. Haemost. , vol.99 , pp. 215-222
    • Jakubowski, J.A.1
  • 47
    • 79952557680 scopus 로고    scopus 로고
    • Monitoring of antiplatelet therapy. Current limitations, challenges, and perspectives
    • Seidel, H., Rahman, M.M. & Scharf R.E. Monitoring of antiplatelet therapy. Current limitations, challenges, and perspectives. Hamostaseologie 31, 41–51 (2011).
    • (2011) Hamostaseologie , vol.31 , pp. 41-51
    • Seidel, H.1    Rahman, M.M.2    Scharf, R.E.3
  • 48
    • 44949173754 scopus 로고    scopus 로고
    • Prognostic significance of post-clopidogrel platelet reactivity assessed by a point-of-care assay on thrombotic events after drug-eluting stent implantation
    • Price, M.J. et al. Prognostic significance of post-clopidogrel platelet reactivity assessed by a point-of-care assay on thrombotic events after drug-eluting stent implantation. Eur. Heart J. 29, 992–1000 (2008).
    • (2008) Eur. Heart J. , vol.29 , pp. 992-1000
    • Price, M.J.1
  • 49
    • 59849120163 scopus 로고    scopus 로고
    • Cardiovascular death and nonfatal myocardial infarction in acute coronary syndrome patients receiving coronary stenting are predicted by residual platelet reactivity to ADP detected by a point-of-care assay: a 12-month follow-up
    • Marcucci, R. et al. Cardiovascular death and nonfatal myocardial infarction in acute coronary syndrome patients receiving coronary stenting are predicted by residual platelet reactivity to ADP detected by a point-of-care assay: a 12-month follow-up. Circulation 119, 237–242 (2009).
    • (2009) Circulation , vol.119 , pp. 237-242
    • Marcucci, R.1
  • 50
    • 84872727527 scopus 로고    scopus 로고
    • Intraindividual and interindividual variabilities in endogenous cortisol 6β-hydroxylation clearance as an index for in vivo CYP3A phenotyping in humans
    • Shibasaki, H. et al. Intraindividual and interindividual variabilities in endogenous cortisol 6β-hydroxylation clearance as an index for in vivo CYP3A phenotyping in humans. Drug Metab. Dispos. 41, 475–479 (2013).
    • (2013) Drug Metab. Dispos. , vol.41 , pp. 475-479
    • Shibasaki, H.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.