메뉴 건너뛰기




Volumn 32, Issue 12, 2015, Pages 3785-3802

Predicting Clearance Mechanism in Drug Discovery: Extended Clearance Classification System (ECCS)

Author keywords

extended clearance classification system (ECCS); hepatic uptake; metabolism; permeability; renal clearance

Indexed keywords

AMPHOLYTE; DRUG;

EID: 84946478501     PISSN: 07248741     EISSN: 1573904X     Source Type: Journal    
DOI: 10.1007/s11095-015-1749-4     Document Type: Review
Times cited : (238)

References (180)
  • 1
    • 79955601786 scopus 로고    scopus 로고
    • Trial watch: Phase II failures: 2008-2010
    • 1:CAS:528:DC%2BC3MXlsVGlsbk%3D 21532551
    • Arrowsmith J. Trial watch: phase II failures: 2008-2010. Nat Rev Drug Discov. 2011;10:328-9.
    • (2011) Nat Rev Drug Discov , vol.10 , pp. 328-329
    • Arrowsmith, J.1
  • 2
    • 84860690620 scopus 로고    scopus 로고
    • Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving Phase II survival
    • 1:CAS:528:DC%2BC38Xmslymurk%3D 22227532
    • Morgan P, Van Der Graaf PH, Arrowsmith J, Feltner DE, Drummond KS, Wegner CD, et al. Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving Phase II survival. Drug Discov Today. 2012;17:419-24.
    • (2012) Drug Discov Today , vol.17 , pp. 419-424
    • Morgan, P.1    Van Der Graaf, P.H.2    Arrowsmith, J.3    Feltner, D.E.4    Drummond, K.S.5    Wegner, C.D.6
  • 3
    • 33847081694 scopus 로고    scopus 로고
    • What is the objective of the mass balance study? A retrospective analysis of data in animal and human excretion studies employing radiolabeled drugs
    • 1:CAS:528:DC%2BD2sXjsF2nsbk%3D 17364879
    • Roffey SJ, Obach RS, Gedge JI, Smith DA. What is the objective of the mass balance study? A retrospective analysis of data in animal and human excretion studies employing radiolabeled drugs. Drug Metab Rev. 2007;39:17-43.
    • (2007) Drug Metab Rev , vol.39 , pp. 17-43
    • Roffey, S.J.1    Obach, R.S.2    Gedge, J.I.3    Smith, D.A.4
  • 4
    • 84888632710 scopus 로고    scopus 로고
    • A perspective on the prediction of drug pharmacokinetics and disposition in drug research and development
    • 1:CAS:528:DC%2BC3sXhvVCjtbrF
    • Di L, Feng B, Goosen TC, Lai Y, Steyn SJ, Varma MV, et al. A perspective on the prediction of drug pharmacokinetics and disposition in drug research and development. Drug Metab Dispose Biol Fate Chem. 2013;41:1975-93.
    • (2013) Drug Metab Dispose Biol Fate Chem , vol.41 , pp. 1975-1993
    • Di, L.1    Feng, B.2    Goosen, T.C.3    Lai, Y.4    Steyn, S.J.5    Varma, M.V.6
  • 5
    • 84875160515 scopus 로고    scopus 로고
    • Model-based approaches to predict drug-drug interactions associated with hepatic uptake transporters: Preclinical, clinical and beyond
    • 1:CAS:528:DC%2BC3sXktFOgsr8%3D 23331046
    • Barton HA, Lai Y, Goosen TC, Jones HM, El-Kattan AF, Gosset JR, et al. Model-based approaches to predict drug-drug interactions associated with hepatic uptake transporters: preclinical, clinical and beyond. Expert Opin Drug Metab Toxicol. 2013;9:459-72.
    • (2013) Expert Opin Drug Metab Toxicol , vol.9 , pp. 459-472
    • Barton, H.A.1    Lai, Y.2    Goosen, T.C.3    Jones, H.M.4    El-Kattan, A.F.5    Gosset, J.R.6
  • 6
    • 84867873811 scopus 로고    scopus 로고
    • Understanding the critical disposition pathways of statins to assess drug-drug interaction risk during drug development: It's not just about OATP1B1
    • 1:CAS:528:DC%2BC38XhsFChsbjL 23047648
    • Elsby R, Hilgendorf C, Fenner K. Understanding the critical disposition pathways of statins to assess drug-drug interaction risk during drug development: it's not just about OATP1B1. Clin Pharmacol Ther. 2012;92:584-98.
    • (2012) Clin Pharmacol Ther , vol.92 , pp. 584-598
    • Elsby, R.1    Hilgendorf, C.2    Fenner, K.3
  • 7
    • 84862076531 scopus 로고    scopus 로고
    • Impact of drug transporter pharmacogenomics on pharmacokinetic and pharmacodynamic variability - Considerations for drug development
    • 1:CAS:528:DC%2BC38XntlOmu70%3D 22509796
    • Lai Y, Varma M, Feng B, Stephens JC, Kimoto E, El-Kattan A, et al. Impact of drug transporter pharmacogenomics on pharmacokinetic and pharmacodynamic variability - considerations for drug development. Expert Opin Drug Metab Toxicol. 2012;8:723-43.
    • (2012) Expert Opin Drug Metab Toxicol , vol.8 , pp. 723-743
    • Lai, Y.1    Varma, M.2    Feng, B.3    Stephens, J.C.4    Kimoto, E.5    El-Kattan, A.6
  • 8
    • 71949127231 scopus 로고    scopus 로고
    • Interplay of transporters and enzymes in drug and metabolite processing
    • 1:CAS:528:DC%2BD1MXhsFShsrzP 19891494
    • Pang KS, Maeng HJ, Fan J. Interplay of transporters and enzymes in drug and metabolite processing. Mol Pharm. 2009;6:1734-55.
    • (2009) Mol Pharm , vol.6 , pp. 1734-1755
    • Pang, K.S.1    Maeng, H.J.2    Fan, J.3
  • 9
    • 33644847684 scopus 로고    scopus 로고
    • Transporters as a determinant of drug clearance and tissue distribution
    • 1:CAS:528:DC%2BD28Xit1Chtrc%3D 16488580
    • Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci. 2006;27:425-46.
    • (2006) Eur J Pharm Sci , vol.27 , pp. 425-446
    • Shitara, Y.1    Horie, T.2    Sugiyama, Y.3
  • 10
    • 11144276585 scopus 로고    scopus 로고
    • The roles of transporters and enzymes in hepatic drug processing
    • Liu L, Pang KS. The roles of transporters and enzymes in hepatic drug processing. Drug Metab Dispose Biol Fate Chem. 2005;33:1-9.
    • (2005) Drug Metab Dispose Biol Fate Chem , vol.33 , pp. 1-9
    • Liu, L.1    Pang, K.S.2
  • 11
    • 84905013159 scopus 로고    scopus 로고
    • Prediction of pharmacokinetics and drug-drug interactions when hepatic transporters are involved
    • 25056496
    • Li R, Barton HA, Varma MV. Prediction of pharmacokinetics and drug-drug interactions when hepatic transporters are involved. Clin Pharmacokinet. 2014;53:659-78.
    • (2014) Clin Pharmacokinet , vol.53 , pp. 659-678
    • Li, R.1    Barton, H.A.2    Varma, M.V.3
  • 12
    • 80052968100 scopus 로고    scopus 로고
    • Identification of the rate-determining process in the hepatic clearance of atorvastatin in a clinical cassette microdosing study
    • 1:CAS:528:DC%2BC3MXht1WqtbbM 21832990
    • Maeda K, Ikeda Y, Fujita T, Yoshida K, Azuma Y, Haruyama Y, et al. Identification of the rate-determining process in the hepatic clearance of atorvastatin in a clinical cassette microdosing study. Clin Pharmacol Ther. 2011;90:575-81.
    • (2011) Clin Pharmacol Ther , vol.90 , pp. 575-581
    • Maeda, K.1    Ikeda, Y.2    Fujita, T.3    Yoshida, K.4    Azuma, Y.5    Haruyama, Y.6
  • 13
    • 0017603437 scopus 로고
    • Hepatic clearance of drugs. I. Theoretical considerations of a "well-stirred" model and a "parallel tube" model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance
    • 1:CAS:528:DyaE1cXhtFGjtLs%3D 599411
    • Pang KS, Rowland M. Hepatic clearance of drugs. I. Theoretical considerations of a "well-stirred" model and a "parallel tube" model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J Pharmacokinet Biopharm. 1977;5:625-53.
    • (1977) J Pharmacokinet Biopharm , vol.5 , pp. 625-653
    • Pang, K.S.1    Rowland, M.2
  • 14
    • 84872234822 scopus 로고    scopus 로고
    • Hepatic and intestinal drug transporters: Prediction of pharmacokinetic effects caused by drug-drug interactions and genetic polymorphisms
    • 1:CAS:528:DC%2BC3sXjt1Wgu78%3D 23140240
    • Yoshida K, Maeda K, Sugiyama Y. Hepatic and intestinal drug transporters: prediction of pharmacokinetic effects caused by drug-drug interactions and genetic polymorphisms. Annu Rev Pharmacol Toxicol. 2013;53:581-612.
    • (2013) Annu Rev Pharmacol Toxicol , vol.53 , pp. 581-612
    • Yoshida, K.1    Maeda, K.2    Sugiyama, Y.3
  • 15
    • 0029926676 scopus 로고    scopus 로고
    • Recent advances in carrier-mediated hepatic uptake and biliary excretion of xenobiotics
    • 1:CAS:528:DyaK28Xit12rurw%3D 8710738
    • Yamazaki M, Suzuki H, Sugiyama Y. Recent advances in carrier-mediated hepatic uptake and biliary excretion of xenobiotics. Pharm Res. 1996;13:497-513.
    • (1996) Pharm Res , vol.13 , pp. 497-513
    • Yamazaki, M.1    Suzuki, H.2    Sugiyama, Y.3
  • 16
    • 0031430085 scopus 로고    scopus 로고
    • Organ clearance concepts: New perspectives on old principles
    • 1:CAS:528:DyaK1cXis1OjsLY%3D 9561488
    • Sirianni GL, Pang KS. Organ clearance concepts: new perspectives on old principles. J Pharmacokinet Biopharm. 1997;25:449-70.
    • (1997) J Pharmacokinet Biopharm , vol.25 , pp. 449-470
    • Sirianni, G.L.1    Pang, K.S.2
  • 17
    • 33748042671 scopus 로고    scopus 로고
    • Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: Drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions
    • 1:CAS:528:DC%2BD28XptFCit7g%3D 16714062
    • Shitara Y, Sugiyama Y. Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions. Pharmacol Ther. 2006;112:71-105.
    • (2006) Pharmacol Ther , vol.112 , pp. 71-105
    • Shitara, Y.1    Sugiyama, Y.2
  • 18
    • 84861234645 scopus 로고    scopus 로고
    • Predicting human hepatic clearance from in vitro drug metabolism and transport data: A scientific and pharmaceutical perspective for assessing drug-drug interactions
    • 1:CAS:528:DC%2BC38XlslShs7s%3D 22407504
    • Camenisch G, Umehara K. Predicting human hepatic clearance from in vitro drug metabolism and transport data: a scientific and pharmaceutical perspective for assessing drug-drug interactions. Biopharm Drug Dispos. 2012;33:179-94.
    • (2012) Biopharm Drug Dispos , vol.33 , pp. 179-194
    • Camenisch, G.1    Umehara, K.2
  • 19
    • 84876752739 scopus 로고    scopus 로고
    • Quantitative prediction of repaglinide-rifampicin complex drug interactions using dynamic and static mechanistic models: Delineating differential CYP3A4 induction and OATP1B1 inhibition potential of rifampicin
    • 1:CAS:528:DC%2BC3sXmvFSmsrY%3D
    • Varma MV, Lin J, Bi YA, Rotter CJ, Fahmi OA, Lam JL, et al. Quantitative prediction of repaglinide-rifampicin complex drug interactions using dynamic and static mechanistic models: delineating differential CYP3A4 induction and OATP1B1 inhibition potential of rifampicin. Drug Metab Dispose Biol Fate Chem. 2013;41:966-74.
    • (2013) Drug Metab Dispose Biol Fate Chem , vol.41 , pp. 966-974
    • Varma, M.V.1    Lin, J.2    Bi, Y.A.3    Rotter, C.J.4    Fahmi, O.A.5    Lam, J.L.6
  • 20
    • 84978238788 scopus 로고    scopus 로고
    • The extended clearance model and its use for the interpretation of hepatobiliary elimination data
    • Camenisch G, Riede J, Kunze A, Huwyler J, Poller B, Umehara K. The extended clearance model and its use for the interpretation of hepatobiliary elimination data. ADMET DMPK. 2015;3:1-14.
    • (2015) ADMET DMPK , vol.3 , pp. 1-14
    • Camenisch, G.1    Riede, J.2    Kunze, A.3    Huwyler, J.4    Poller, B.5    Umehara, K.6
  • 21
    • 34748925493 scopus 로고    scopus 로고
    • Prediction of human pharmacokinetics using physiologically based modeling: A retrospective analysis of 26 clinically tested drugs
    • De Buck SS, Sinha VK, Fenu LA, Nijsen MJ, Mackie CE, Gilissen RA. Prediction of human pharmacokinetics using physiologically based modeling: a retrospective analysis of 26 clinically tested drugs. Drug Metab Dispose Biol Fate Chem. 2007;35:1766-80.
    • (2007) Drug Metab Dispose Biol Fate Chem , vol.35 , pp. 1766-1780
    • De Buck, S.S.1    Sinha, V.K.2    Fenu, L.A.3    Nijsen, M.J.4    MacKie, C.E.5    Gilissen, R.A.6
  • 22
    • 79953296456 scopus 로고    scopus 로고
    • Simulation of human intravenous and oral pharmacokinetics of 21 diverse compounds using physiologically based pharmacokinetic modelling
    • 1:CAS:528:DC%2BC3MXntlKlsbc%3D 21456633
    • Jones HM, Gardner IB, Collard WT, Stanley PJ, Oxley P, Hosea NA, et al. Simulation of human intravenous and oral pharmacokinetics of 21 diverse compounds using physiologically based pharmacokinetic modelling. Clin Pharmacokinet. 2011;50:331-47.
    • (2011) Clin Pharmacokinet , vol.50 , pp. 331-347
    • Jones, H.M.1    Gardner, I.B.2    Collard, W.T.3    Stanley, P.J.4    Oxley, P.5    Hosea, N.A.6
  • 23
    • 0030799001 scopus 로고    scopus 로고
    • The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data
    • 1:CAS:528:DyaK2sXmslCltr0%3D 9336307
    • Obach RS, Baxter JG, Liston TE, Silber BM, Jones BC, Macintyre F, et al. The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J Pharmacol Exp Ther. 1997;283:46-58.
    • (1997) J Pharmacol Exp Ther , vol.283 , pp. 46-58
    • Obach, R.S.1    Baxter, J.G.2    Liston, T.E.3    Silber, B.M.4    Jones, B.C.5    Macintyre, F.6
  • 24
    • 6944221357 scopus 로고    scopus 로고
    • Drug-drug interactions for UDP-glucuronosyltransferase substrates: A pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios
    • 1:CAS:528:DC%2BD2cXovFCktLg%3D 15304429
    • Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, et al. Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos. 2004;32:1201-8.
    • (2004) Drug Metab Dispos , vol.32 , pp. 1201-1208
    • Williams, J.A.1    Hyland, R.2    Jones, B.C.3    Smith, D.A.4    Hurst, S.5    Goosen, T.C.6
  • 26
    • 68149170038 scopus 로고    scopus 로고
    • The role of transporters in the pharmacokinetics of orally administered drugs
    • 2719753 1:CAS:528:DC%2BD1MXnvFKgu7w%3D 19568696
    • Shugarts S, Bene LZ. The role of transporters in the pharmacokinetics of orally administered drugs. Pharm Res. 2009;26:2039-54.
    • (2009) Pharm Res , vol.26 , pp. 2039-2054
    • Shugarts, S.1    Bene, L.Z.2
  • 27
    • 84872229531 scopus 로고    scopus 로고
    • Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: Their roles in hepatic clearance and intestinal absorption
    • 1:CAS:528:DC%2BC3sXhtFSls7Y%3D 23115084
    • Shitara Y, Maeda K, Ikejiri K, Yoshida K, Horie T, Sugiyama Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34:45-78.
    • (2013) Biopharm Drug Dispos , vol.34 , pp. 45-78
    • Shitara, Y.1    Maeda, K.2    Ikejiri, K.3    Yoshida, K.4    Horie, T.5    Sugiyama, Y.6
  • 28
    • 82955168418 scopus 로고    scopus 로고
    • The evolution of the OATP hepatic uptake transport protein family in DMPK sciences: From obscure liver transporters to key determinants of hepatobiliary clearance
    • 1:CAS:528:DC%2BC3MXhsFKgtbvK 22077101
    • Fenner KS, Jones HM, Ullah M, Kempshall S, Dickins M, Lai Y, et al. The evolution of the OATP hepatic uptake transport protein family in DMPK sciences: from obscure liver transporters to key determinants of hepatobiliary clearance. Xenobiotica. 2012;42:28-45.
    • (2012) Xenobiotica , vol.42 , pp. 28-45
    • Fenner, K.S.1    Jones, H.M.2    Ullah, M.3    Kempshall, S.4    Dickins, M.5    Lai, Y.6
  • 29
    • 70350048926 scopus 로고    scopus 로고
    • Impact of OATP transporters on pharmacokinetics
    • 2765590 1:CAS:528:DC%2BD1MXhsVSks7rE 19785645
    • Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharmacol. 2009;158:693-705.
    • (2009) Br J Pharmacol , vol.158 , pp. 693-705
    • Kalliokoski, A.1    Niemi, M.2
  • 30
    • 84863999189 scopus 로고    scopus 로고
    • Whole-body distribution and radiation dosimetry of [11C]telmisartan as a biomarker for hepatic organic anion transporting polypeptide (OATP) 1B3
    • 1:CAS:528:DC%2BC38XjvVWls7c%3D 22421430
    • Shimizu K, Takashima T, Yamane T, Sasaki M, Kageyama H, Hashizume Y, et al. Whole-body distribution and radiation dosimetry of [11C]telmisartan as a biomarker for hepatic organic anion transporting polypeptide (OATP) 1B3. Nucl Med Biol. 2012;39:847-53.
    • (2012) Nucl Med Biol , vol.39 , pp. 847-853
    • Shimizu, K.1    Takashima, T.2    Yamane, T.3    Sasaki, M.4    Kageyama, H.5    Hashizume, Y.6
  • 31
    • 33750192677 scopus 로고    scopus 로고
    • Biliary secretion of rosuvastatin and bile acids in humans during the absorption phase
    • 1:CAS:528:DC%2BD28XhtFentrzI 16806856
    • Bergman E, Forsell P, Tevell A, Persson EM, Hedeland M, Bondesson U, et al. Biliary secretion of rosuvastatin and bile acids in humans during the absorption phase. Eur J Pharm Sci. 2006;29:205-14.
    • (2006) Eur J Pharm Sci , vol.29 , pp. 205-214
    • Bergman, E.1    Forsell, P.2    Tevell, A.3    Persson, E.M.4    Hedeland, M.5    Bondesson, U.6
  • 32
    • 0029782268 scopus 로고    scopus 로고
    • Primary active transport of pravastatin across the liver canalicular membrane in normal and mutant Eisai hyperbilirubinemic rats
    • 1:CAS:528:DyaK28XmtFSlu7k%3D 8894118
    • Yamazaki M, Kobayashi K, Sugiyama Y. Primary active transport of pravastatin across the liver canalicular membrane in normal and mutant Eisai hyperbilirubinemic rats. Biopharm Drug Dispos. 1996;17:607-21.
    • (1996) Biopharm Drug Dispos , vol.17 , pp. 607-621
    • Yamazaki, M.1    Kobayashi, K.2    Sugiyama, Y.3
  • 33
    • 84866708911 scopus 로고    scopus 로고
    • Physiologically based modeling of pravastatin transporter-mediated hepatobiliary disposition and drug-drug interactions.
    • Varma MV, Lai Y, Feng B, Litchfield J, Goosen TC, Bergman A. Physiologically based modeling of pravastatin transporter-mediated hepatobiliary disposition and drug-drug interactions. Pharm Res. 2012.
    • (2012) Pharm Res.
    • Varma, M.V.1    Lai, Y.2    Feng, B.3    Litchfield, J.4    Goosen, T.C.5    Bergman, A.6
  • 34
    • 59649130318 scopus 로고    scopus 로고
    • Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans
    • 1:CAS:528:DC%2BD1MXhs1Kgs7w%3D 19001154
    • Watanabe T, Kusuhara H, Maeda K, Shitara Y, Sugiyama Y. Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans. J Pharmacol Exp Ther. 2009;328:652-62.
    • (2009) J Pharmacol Exp Ther , vol.328 , pp. 652-662
    • Watanabe, T.1    Kusuhara, H.2    Maeda, K.3    Shitara, Y.4    Sugiyama, Y.5
  • 35
    • 33846571368 scopus 로고    scopus 로고
    • Effect of OATP1B transporter inhibition on the pharmacokinetics of atorvastatin in healthy volunteers
    • 17192770
    • Lau Y, Huang Y, Frassetto L, Benet L. Effect of OATP1B transporter inhibition on the pharmacokinetics of atorvastatin in healthy volunteers. Clin Pharmacol Ther. 2006;81:194-204.
    • (2006) Clin Pharmacol Ther , vol.81 , pp. 194-204
    • Lau, Y.1    Huang, Y.2    Frassetto, L.3    Benet, L.4
  • 36
    • 84903633472 scopus 로고    scopus 로고
    • Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide
    • 4070267 1:CAS:528:DC%2BC2cXosVWgtr8%3D 24839071
    • Varma MV, Scialis RJ, Lin J, Bi YA, Rotter CJ, Goosen TC, et al. Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide. AAPS J. 2014;16:736-48.
    • (2014) AAPS J , vol.16 , pp. 736-748
    • Varma, M.V.1    Scialis, R.J.2    Lin, J.3    Bi, Y.A.4    Rotter, C.J.5    Goosen, T.C.6
  • 37
    • 57749171180 scopus 로고    scopus 로고
    • Elucidating rifampin's inducing and inhibiting effects on glyburide pharmacokinetics and blood glucose in healthy volunteers: Unmasking the differential effects of enzyme induction and transporter inhibition for a drug and its primary metabolite
    • 3582657 18843263
    • Zheng H, Huang Y, Frassetto L, Benet L. Elucidating rifampin's inducing and inhibiting effects on glyburide pharmacokinetics and blood glucose in healthy volunteers: unmasking the differential effects of enzyme induction and transporter inhibition for a drug and its primary metabolite. Clin Pharmacol Ther. 2008;85:78-85.
    • (2008) Clin Pharmacol Ther , vol.85 , pp. 78-85
    • Zheng, H.1    Huang, Y.2    Frassetto, L.3    Benet, L.4
  • 38
    • 56149107690 scopus 로고    scopus 로고
    • Effects of gemfibrozil and atorvastatin on the pharmacokinetics of repaglinide in relation to SLCO1B1 polymorphism
    • 1:CAS:528:DC%2BD1cXhtFeitbvF 19238654
    • Kalliokoski A, Backman JT, Kurkinen KJ, Neuvonen PJ, Niemi M. Effects of gemfibrozil and atorvastatin on the pharmacokinetics of repaglinide in relation to SLCO1B1 polymorphism. Clin Pharmacol Ther. 2008;84:488-96.
    • (2008) Clin Pharmacol Ther , vol.84 , pp. 488-496
    • Kalliokoski, A.1    Backman, J.T.2    Kurkinen, K.J.3    Neuvonen, P.J.4    Niemi, M.5
  • 39
    • 84876483882 scopus 로고    scopus 로고
    • Mechanistic modeling to predict the transporter- and enzyme-mediated drug-drug interactions of repaglinide
    • 1:CAS:528:DC%2BC3sXlvVKjug%3D%3D 23307347
    • Varma MV, Lai Y, Kimoto E, Goosen TC, El-Kattan AF, Kumar V. Mechanistic modeling to predict the transporter- and enzyme-mediated drug-drug interactions of repaglinide. Pharm Res. 2013;30:1188-99.
    • (2013) Pharm Res , vol.30 , pp. 1188-1199
    • Varma, M.V.1    Lai, Y.2    Kimoto, E.3    Goosen, T.C.4    El-Kattan, A.F.5    Kumar, V.6
  • 41
    • 49949104757 scopus 로고    scopus 로고
    • SLCO1B1 variants and statin-induced myopathy - A genomewide study
    • Group SC, Link E, Parish S, Armitage J, Bowman L, Heath S, et al. SLCO1B1 variants and statin-induced myopathy - a genomewide study. N Engl J Med. 2008;359:789-99.
    • (2008) N Engl J Med , vol.359 , pp. 789-799
    • Link, E.1    Parish, S.2    Armitage, J.3    Bowman, L.4    Heath, S.5
  • 42
    • 67650753944 scopus 로고    scopus 로고
    • Genetic polymorphisms of uptake (OATP1B1, 1B3) and efflux (MRP2, BCRP) transporters: Implications for inter-individual differences in the pharmacokinetics and pharmacodynamics of statins and other clinically relevant drugs
    • 1:CAS:528:DC%2BD1MXnsFamsL0%3D
    • Ieiri I, Higuchi S, Sugiyama Y. Genetic polymorphisms of uptake (OATP1B1, 1B3) and efflux (MRP2, BCRP) transporters: implications for inter-individual differences in the pharmacokinetics and pharmacodynamics of statins and other clinically relevant drugs. Exp Opin Drug Metab Toxicol. 2009;5:703-29.
    • (2009) Exp Opin Drug Metab Toxicol , vol.5 , pp. 703-729
    • Ieiri, I.1    Higuchi, S.2    Sugiyama, Y.3
  • 43
    • 18744388590 scopus 로고    scopus 로고
    • Acute effects of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype∗17
    • 1:CAS:528:DC%2BD2MXjslyquro%3D 15864131
    • Niemi M, Neuvonen PJ, Hofmann U, Backman JT, Schwab M, Lutjohann D, et al. Acute effects of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype∗17. Pharmacogenet Genomics. 2005;15:303-9.
    • (2005) Pharmacogenet Genomics , vol.15 , pp. 303-309
    • Niemi, M.1    Neuvonen, P.J.2    Hofmann, U.3    Backman, J.T.4    Schwab, M.5    Lutjohann, D.6
  • 45
    • 84863894140 scopus 로고    scopus 로고
    • Physicochemical property space of hepatobiliary transport and computational models for predicting rat biliary excretion
    • 1:CAS:528:DC%2BC38XhtF2iurnJ
    • Varma MV, Chang G, Lai Y, Feng B, El-Kattan AF, Litchfield J, et al. Physicochemical property space of hepatobiliary transport and computational models for predicting rat biliary excretion. Drug Metab Dis Biol Fate Chem. 2012;40:1527-37.
    • (2012) Drug Metab Dis Biol Fate Chem , vol.40 , pp. 1527-1537
    • Varma, M.V.1    Chang, G.2    Lai, Y.3    Feng, B.4    El-Kattan, A.F.5    Litchfield, J.6
  • 46
    • 77954355830 scopus 로고    scopus 로고
    • Pharmacokinetic modeling of the hepatobiliary transport mediated by cooperation of uptake and efflux transporters
    • 1:CAS:528:DC%2BC3cXotlSmt7o%3D 20175646
    • Kusuhara H, Sugiyama Y. Pharmacokinetic modeling of the hepatobiliary transport mediated by cooperation of uptake and efflux transporters. Drug Metab Rev. 2010;42:539-50.
    • (2010) Drug Metab Rev , vol.42 , pp. 539-550
    • Kusuhara, H.1    Sugiyama, Y.2
  • 47
    • 70349399011 scopus 로고    scopus 로고
    • Identification of interspecies difference in hepatobiliary transporters to improve extrapolation of human biliary secretion
    • 1:CAS:528:DC%2BD1MXhtFCgsrfP
    • Lai Y. Identification of interspecies difference in hepatobiliary transporters to improve extrapolation of human biliary secretion. Exp Opin Drug Metab Toxicol. 2009;5:1175-87.
    • (2009) Exp Opin Drug Metab Toxicol , vol.5 , pp. 1175-1187
    • Lai, Y.1
  • 48
    • 33745231761 scopus 로고    scopus 로고
    • Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II AT1-receptor, in humans
    • 1:CAS:528:DC%2BD28XmsVKqsbw%3D
    • Yamashiro W, Maeda K, Hirouchi M, Adachi Y, Hu Z, Sugiyama Y. Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II AT1-receptor, in humans. Drug Metab Dis Biol Fate Chem. 2006;34:1247-54.
    • (2006) Drug Metab Dis Biol Fate Chem , vol.34 , pp. 1247-1254
    • Yamashiro, W.1    Maeda, K.2    Hirouchi, M.3    Adachi, Y.4    Hu, Z.5    Sugiyama, Y.6
  • 49
    • 72349089654 scopus 로고    scopus 로고
    • Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data
    • 1:CAS:528:DC%2BD1MXhsV2jt7zL 19936896
    • Poirier A, Cascais A-C, Funk C, Lavé T. Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data. J Pharmacokinet Pharmacodyn. 2009;36:585-611.
    • (2009) J Pharmacokinet Pharmacodyn , vol.36 , pp. 585-611
    • Poirier, A.1    Cascais, A.-C.2    Funk, C.3    Lavé, T.4
  • 50
    • 73349114878 scopus 로고    scopus 로고
    • Prediction of biliary excretion in rats and humans using molecular weight and quantitative structure-pharmacokinetic relationships
    • 2758117 1:CAS:528:DC%2BD1MXhsFGmur7J 19593675
    • Yang X, Gandhi YA, Duignan DB, Morris ME. Prediction of biliary excretion in rats and humans using molecular weight and quantitative structure-pharmacokinetic relationships. AAPS J. 2009;11:511-25.
    • (2009) AAPS J , vol.11 , pp. 511-525
    • Yang, X.1    Gandhi, Y.A.2    Duignan, D.B.3    Morris, M.E.4
  • 51
    • 42549095582 scopus 로고    scopus 로고
    • Prediction of human pharmacokinetics-biliary and intestinal clearance and enterohepatic circulation
    • 1:CAS:528:DC%2BD1cXltlemsbc%3D 18416932
    • Fagerholm U. Prediction of human pharmacokinetics-biliary and intestinal clearance and enterohepatic circulation. J Pharm Pharmacol. 2008;60:535-42.
    • (2008) J Pharm Pharmacol , vol.60 , pp. 535-542
    • Fagerholm, U.1
  • 52
    • 76749103891 scopus 로고    scopus 로고
    • In silico prediction of biliary excretion of drugs in rats based on physicochemical properties
    • 1:CAS:528:DC%2BC3cXjt1Ort7g%3D
    • Luo G, Johnson S, Hsueh MM, Zheng J, Cai H, Xin B, et al. In silico prediction of biliary excretion of drugs in rats based on physicochemical properties. Drug Metab Dis Biol Fate Chem. 2010;38:422-30.
    • (2010) Drug Metab Dis Biol Fate Chem , vol.38 , pp. 422-430
    • Luo, G.1    Johnson, S.2    Hsueh, M.M.3    Zheng, J.4    Cai, H.5    Xin, B.6
  • 53
    • 0013597612 scopus 로고
    • Biliary excretion in foreign compounds. Sulphonamide drugs in the rat
    • 1198452 1:CAS:528:DyaF1cXitVagtA%3D%3D 16742557
    • Millburn P, Smith RL, Williams RT. Biliary excretion in foreign compounds. Sulphonamide drugs in the rat. Biochem J. 1967;105:1283-7.
    • (1967) Biochem J , vol.105 , pp. 1283-1287
    • Millburn, P.1    Smith, R.L.2    Williams, R.T.3
  • 54
    • 0013597612 scopus 로고
    • Biliary excretion of foreign compounds. Biphenyl, stilboestrol and phenolphthalein in the rat: Molecular weight, polarity and metabolism as factors in biliary excretion
    • 1198451 1:CAS:528:DyaF1cXitVagtw%3D%3D 16742556
    • Millburn P, Smith RL, Williams RT. Biliary excretion of foreign compounds. Biphenyl, stilboestrol and phenolphthalein in the rat: molecular weight, polarity and metabolism as factors in biliary excretion. Biochem J. 1967;105:1275-81.
    • (1967) Biochem J , vol.105 , pp. 1275-1281
    • Millburn, P.1    Smith, R.L.2    Williams, R.T.3
  • 55
    • 44149128448 scopus 로고    scopus 로고
    • Involvement of multidrug resistance-associated protein 2 (Abcc2) in molecular weight-dependent biliary excretion of beta-lactam antibiotics
    • 1:CAS:528:DC%2BD1cXmsVOmurw%3D
    • Kato Y, Takahara S, Kato S, Kubo Y, Sai Y, Tamai I, et al. Involvement of multidrug resistance-associated protein 2 (Abcc2) in molecular weight-dependent biliary excretion of beta-lactam antibiotics. Drug Metab Dis Biol Fate Chem. 2008;36:1088-96.
    • (2008) Drug Metab Dis Biol Fate Chem , vol.36 , pp. 1088-1096
    • Kato, Y.1    Takahara, S.2    Kato, S.3    Kubo, Y.4    Sai, Y.5    Tamai, I.6
  • 56
    • 63449139456 scopus 로고    scopus 로고
    • Structure of P-glycoprotein reveals a molecular basis for poly-specific drug binding
    • 2720052 1:CAS:528:DC%2BD1MXjs1Kgtro%3D 19325113
    • Aller SG, Yu J, Ward A, Weng Y, Chittaboina S, Zhuo R, et al. Structure of P-glycoprotein reveals a molecular basis for poly-specific drug binding. Science. 2009;323:1718-22.
    • (2009) Science , vol.323 , pp. 1718-1722
    • Aller, S.G.1    Yu, J.2    Ward, A.3    Weng, Y.4    Chittaboina, S.5    Zhuo, R.6
  • 57
    • 73349137921 scopus 로고    scopus 로고
    • Structure-activity relationships and quantitative structure-activity relationships for breast cancer resistance protein (ABCG2)
    • 2758125 1:CAS:528:DC%2BD1MXhsFGmur7E 19629710
    • Gandhi YA, Morris ME. Structure-activity relationships and quantitative structure-activity relationships for breast cancer resistance protein (ABCG2). AAPS J. 2009;11:541-52.
    • (2009) AAPS J , vol.11 , pp. 541-552
    • Gandhi, Y.A.1    Morris, M.E.2
  • 58
    • 67650084777 scopus 로고    scopus 로고
    • Identification of novel specific and general inhibitors of the three major human ATP-binding cassette transporters P-gp, BCRP and MRP2 among registered drugs
    • 1:CAS:528:DC%2BD1MXlsFOmt70%3D 19421845
    • Matsson P, Pedersen JM, Norinder U, Bergstrom CA, Artursson P. Identification of novel specific and general inhibitors of the three major human ATP-binding cassette transporters P-gp, BCRP and MRP2 among registered drugs. Pharm Res. 2009;26:1816-31.
    • (2009) Pharm Res , vol.26 , pp. 1816-1831
    • Matsson, P.1    Pedersen, J.M.2    Norinder, U.3    Bergstrom, C.A.4    Artursson, P.5
  • 59
    • 79851494257 scopus 로고    scopus 로고
    • Targeting intestinal transporters for optimizing oral drug absorption
    • 1:CAS:528:DC%2BC3MXhsl2kt7c%3D 21189135
    • Varma MV, Ambler CM, Ullah M, Rotter CJ, Sun H, Litchfield J, et al. Targeting intestinal transporters for optimizing oral drug absorption. Curr Drug Metab. 2010;11:730-42.
    • (2010) Curr Drug Metab , vol.11 , pp. 730-742
    • Varma, M.V.1    Ambler, C.M.2    Ullah, M.3    Rotter, C.J.4    Sun, H.5    Litchfield, J.6
  • 60
    • 33144466513 scopus 로고    scopus 로고
    • PH-dependent functional activity of P-glycoprotein in limiting intestinal absorption of protic drugs: Kinetic analysis of quinidine efflux in situ
    • 1:CAS:528:DC%2BD2MXht12ktrjO 16258992
    • Varma MV, Panchagnula R. pH-dependent functional activity of P-glycoprotein in limiting intestinal absorption of protic drugs: kinetic analysis of quinidine efflux in situ. J Pharm Sci. 2005;94:2632-43.
    • (2005) J Pharm Sci , vol.94 , pp. 2632-2643
    • Varma, M.V.1    Panchagnula, R.2
  • 61
    • 84860755198 scopus 로고    scopus 로고
    • PH-Dependent solubility and permeability criteria for provisional biopharmaceutics classification (BCS and BDDCS) in early drug discovery
    • 1:CAS:528:DC%2BC38Xlt1Ont74%3D 22489626
    • Varma MV, Gardner I, Steyn SJ, Nkansah P, Rotter CJ, Whitney-Pickett C, et al. pH-Dependent solubility and permeability criteria for provisional biopharmaceutics classification (BCS and BDDCS) in early drug discovery. Mol Pharm. 2012;9:1199-212.
    • (2012) Mol Pharm , vol.9 , pp. 1199-1212
    • Varma, M.V.1    Gardner, I.2    Steyn, S.J.3    Nkansah, P.4    Rotter, C.J.5    Whitney-Pickett, C.6
  • 62
    • 77953290975 scopus 로고    scopus 로고
    • Increased affinity to canalicular P-gp via formation of lipophilic ion-pair complexes with endogenous bile salts is associated with mw threshold in hepatobiliary excretion of quaternary ammonium compounds
    • 1:CAS:528:DC%2BC3cXjtVWrsLY%3D 20221674
    • Song IS, Choi MK, Jin QR, Shim WS, Shim CK. Increased affinity to canalicular P-gp via formation of lipophilic ion-pair complexes with endogenous bile salts is associated with mw threshold in hepatobiliary excretion of quaternary ammonium compounds. Pharm Res. 2010;27:823-31.
    • (2010) Pharm Res , vol.27 , pp. 823-831
    • Song, I.S.1    Choi, M.K.2    Jin, Q.R.3    Shim, W.S.4    Shim, C.K.5
  • 63
    • 83555174958 scopus 로고    scopus 로고
    • BDDCS applied to over 900 drugs
    • 3231854 1:CAS:528:DC%2BC3MXhsFamsbfE 21818695
    • Benet LZ, Broccatelli F, Oprea TI. BDDCS applied to over 900 drugs. AAPS J. 2011;13:519-47.
    • (2011) AAPS J , vol.13 , pp. 519-547
    • Benet, L.Z.1    Broccatelli, F.2    Oprea, T.I.3
  • 64
    • 39149115065 scopus 로고    scopus 로고
    • Predicting drug disposition, absorption/elimination/transporter interplay and the role of food on drug absorption
    • 2292816 1:CAS:528:DC%2BD1cXitVOmsr0%3D 18199522
    • Custodio JM, Wu C-Y, Benet LZ. Predicting drug disposition, absorption/elimination/transporter interplay and the role of food on drug absorption. Adv Drug Deliv Rev. 2008;60:717-33.
    • (2008) Adv Drug Deliv Rev , vol.60 , pp. 717-733
    • Custodio, J.M.1    Wu, C.-Y.2    Benet, L.Z.3
  • 65
    • 17644380257 scopus 로고    scopus 로고
    • Predicting drug disposition via application of BCS: Transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system
    • 1:CAS:528:DC%2BD2MXhtlCks70%3D 15771225
    • Wu C-Y, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res. 2005;22:11-23.
    • (2005) Pharm Res , vol.22 , pp. 11-23
    • Wu, C.-Y.1    Benet, L.Z.2
  • 66
    • 84870249445 scopus 로고    scopus 로고
    • Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings
    • Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2012;64:4-17.
    • (2012) Adv Drug Deliv Rev , vol.64 , pp. 4-17
    • Lipinski, C.A.1    Lombardo, F.2    Dominy, B.W.3    Feeney, P.J.4
  • 67
    • 0037030653 scopus 로고    scopus 로고
    • Molecular properties that influence the oral bioavailability of drug candidates
    • 1:CAS:528:DC%2BD38XjsFCmt7g%3D 12036371
    • Veber DF, Johnson SR, Cheng H-Y, Smith BR, Ward KW, Kopple KD. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem. 2002;45:2615-23.
    • (2002) J Med Chem , vol.45 , pp. 2615-2623
    • Veber, D.F.1    Johnson, S.R.2    Cheng, H.-Y.3    Smith, B.R.4    Ward, K.W.5    Kopple, K.D.6
  • 68
    • 0032189375 scopus 로고    scopus 로고
    • Estimation of permeability by passive diffusion through Caco-2 cell monolayers using the drugs' lipophilicity and molecular weight
    • 1:CAS:528:DyaK1cXmtl2nsr0%3D
    • Camenisch G, Alsenz J, van de Waterbeemd H, Folkers G. Estimation of permeability by passive diffusion through Caco-2 cell monolayers using the drugs' lipophilicity and molecular weight. Eur J Pharm Sci. 1998;6:313-9.
    • (1998) Eur J Pharm Sci , vol.6 , pp. 313-319
    • Camenisch, G.1    Alsenz, J.2    Van De Waterbeemd, H.3    Folkers, G.4
  • 69
    • 77249122159 scopus 로고    scopus 로고
    • Physicochemical space for optimum oral bioavailability: Contribution of human intestinal absorption and first-pass elimination
    • 1:CAS:528:DC%2BC3cXkvFGisg%3D%3D 20070106
    • Varma MV, Obach RS, Rotter C, Miller HR, Chang G, Steyn SJ, et al. Physicochemical space for optimum oral bioavailability: contribution of human intestinal absorption and first-pass elimination. J Med Chem. 2010;53:1098-108.
    • (2010) J Med Chem , vol.53 , pp. 1098-1108
    • Varma, M.V.1    Obach, R.S.2    Rotter, C.3    Miller, H.R.4    Chang, G.5    Steyn, S.J.6
  • 70
    • 46449114275 scopus 로고    scopus 로고
    • Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds
    • 1:CAS:528:DC%2BD1cXotVensr0%3D
    • Obach RS, Lombardo F, Waters NJ. Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds. Drug Metab Dis Biol Fate Chem. 2008;36:1385-405.
    • (2008) Drug Metab Dis Biol Fate Chem , vol.36 , pp. 1385-1405
    • Obach, R.S.1    Lombardo, F.2    Waters, N.J.3
  • 71
    • 0032804701 scopus 로고    scopus 로고
    • Homology modelling of human cytochromes P450 involved in xenobiotic metabolism and rationalization of substrate selectivity
    • 1:CAS:528:DyaK1MXkvVCmsbY%3D 10445400
    • Lewis DF. Homology modelling of human cytochromes P450 involved in xenobiotic metabolism and rationalization of substrate selectivity. Exp Toxicol Pathol. 1999;51:369-74.
    • (1999) Exp Toxicol Pathol , vol.51 , pp. 369-374
    • Lewis, D.F.1
  • 72
    • 0036223228 scopus 로고    scopus 로고
    • Structure-activity relationship for human cytochrome P450 substrates and inhibitors
    • 1:CAS:528:DC%2BD38Xjs1KgsLk%3D 11996013
    • Lewis DF, Modi S, Dickins M. Structure-activity relationship for human cytochrome P450 substrates and inhibitors. Drug Metab Rev. 2002;34:69-82.
    • (2002) Drug Metab Rev , vol.34 , pp. 69-82
    • Lewis, D.F.1    Modi, S.2    Dickins, M.3
  • 73
    • 24344439264 scopus 로고    scopus 로고
    • Transporters and drug therapy: Implications for drug disposition and disease
    • 1:CAS:528:DC%2BD2MXpvV2rt78%3D 16153397
    • Ho RH, Kim RB. Transporters and drug therapy: implications for drug disposition and disease. Clin Pharmacol Ther. 2005;78:260-77.
    • (2005) Clin Pharmacol Ther , vol.78 , pp. 260-277
    • Ho, R.H.1    Kim, R.B.2
  • 75
    • 35848946788 scopus 로고    scopus 로고
    • Prediction of human pharmacokinetics - Renal metabolic and excretion clearance
    • 17976256
    • Fagerholm U. Prediction of human pharmacokinetics - renal metabolic and excretion clearance. J Pharm Pharmacol. 2007;59:1463-71.
    • (2007) J Pharm Pharmacol , vol.59 , pp. 1463-1471
    • Fagerholm, U.1
  • 76
    • 28444444126 scopus 로고    scopus 로고
    • Effects of renal failure on drug transport and metabolism
    • 1:CAS:528:DC%2BD2MXht12ktbbM 16085315
    • Sun H, Frassetto L, Benet LZ. Effects of renal failure on drug transport and metabolism. Pharmacol Ther. 2006;109:1-11.
    • (2006) Pharmacol Ther , vol.109 , pp. 1-11
    • Sun, H.1    Frassetto, L.2    Benet, L.Z.3
  • 77
    • 57749202043 scopus 로고    scopus 로고
    • Transport mechanisms of carnosine in SKPT cells: Contribution of apical and basolateral membrane transporters
    • 2913304 1:CAS:528:DC%2BD1cXhsFSjsr7J 18820998
    • Jappar D, Hu Y, Keep RF, Smith DE. Transport mechanisms of carnosine in SKPT cells: contribution of apical and basolateral membrane transporters. Pharm Res. 2009;26:172-81.
    • (2009) Pharm Res , vol.26 , pp. 172-181
    • Jappar, D.1    Hu, Y.2    Keep, R.F.3    Smith, D.E.4
  • 78
    • 0035209269 scopus 로고    scopus 로고
    • Mechanisms and clinical implications of renal drug excretion
    • 1:CAS:528:DC%2BD38XjvVGqsw%3D%3D 11768771
    • Masereeuw R, Russel FG. Mechanisms and clinical implications of renal drug excretion. Drug Metab Rev. 2001;33:299-351.
    • (2001) Drug Metab Rev , vol.33 , pp. 299-351
    • Masereeuw, R.1    Russel, F.G.2
  • 79
    • 53549119190 scopus 로고    scopus 로고
    • Structural requirements for drug inhibition of the liver specific human organic cation transport protein 1
    • 1:CAS:528:DC%2BD1cXhtFeltL7P 18788725
    • Ahlin G, Karlsson J, Pedersen JM, Gustavsson L, Larsson R, Matsson P, et al. Structural requirements for drug inhibition of the liver specific human organic cation transport protein 1. J Med Chem. 2008;51:5932-42.
    • (2008) J Med Chem , vol.51 , pp. 5932-5942
    • Ahlin, G.1    Karlsson, J.2    Pedersen, J.M.3    Gustavsson, L.4    Larsson, R.5    Matsson, P.6
  • 80
    • 0033849976 scopus 로고    scopus 로고
    • Cellular and molecular aspects of drug transport in the kidney
    • 1:CAS:528:DC%2BD3cXmsleitr4%3D 10972658
    • Inui KI, Masuda S, Saito H. Cellular and molecular aspects of drug transport in the kidney. Kidney Int. 2000;58:944-58.
    • (2000) Kidney Int , vol.58 , pp. 944-958
    • Inui, K.I.1    Masuda, S.2    Saito, H.3
  • 83
    • 0035950120 scopus 로고    scopus 로고
    • Physicochemical determinants of passive membrane permeability: Role of solute hydrogen-bonding potential and volume
    • 1:CAS:528:DC%2BD3MXmvFOmtrs%3D 11606137
    • Goodwin JT, Conradi RA, Ho NF, Burton PS. Physicochemical determinants of passive membrane permeability: role of solute hydrogen-bonding potential and volume. J Med Chem. 2001;44:3721-9.
    • (2001) J Med Chem , vol.44 , pp. 3721-3729
    • Goodwin, J.T.1    Conradi, R.A.2    Ho, N.F.3    Burton, P.S.4
  • 84
    • 14644419054 scopus 로고    scopus 로고
    • Functional role of P-glycoprotein in limiting intestinal absorption of drugs: Contribution of passive permeability to P-glycoprotein mediated efflux transport
    • 1:CAS:528:DC%2BD2cXhtVSmu7vF 15804173
    • Varma MV, Sateesh K, Panchagnula R. Functional role of P-glycoprotein in limiting intestinal absorption of drugs: contribution of passive permeability to P-glycoprotein mediated efflux transport. Mol Pharm. 2005;2:12-21.
    • (2005) Mol Pharm , vol.2 , pp. 12-21
    • Varma, M.V.1    Sateesh, K.2    Panchagnula, R.3
  • 85
    • 0030850803 scopus 로고    scopus 로고
    • Renal transporters for organic anions and organic cations. Structural requirements for substrates
    • 1:CAS:528:DyaK2sXlt1Sitbo%3D 9230087
    • Ullrich KJ. Renal transporters for organic anions and organic cations. Structural requirements for substrates. J Membr Biol. 1997;158:95-107.
    • (1997) J Membr Biol , vol.158 , pp. 95-107
    • Ullrich, K.J.1
  • 86
    • 0344838436 scopus 로고    scopus 로고
    • Influence of molecular structure on substrate binding to the human organic cation transporter, hOCT1
    • 1:CAS:528:DC%2BD3sXitFyrs7s%3D 12606755
    • Bednarczyk D, Ekins S, Wikel JH, Wright SH. Influence of molecular structure on substrate binding to the human organic cation transporter, hOCT1. Mol Pharmacol. 2003;63:489-98.
    • (2003) Mol Pharmacol , vol.63 , pp. 489-498
    • Bednarczyk, D.1    Ekins, S.2    Wikel, J.H.3    Wright, S.H.4
  • 87
    • 77954846109 scopus 로고    scopus 로고
    • Renal clearance in drug discovery and development: Molecular descriptors, drug transporters and disease state
    • 1:CAS:528:DC%2BC3cXptVSqtbc%3D
    • Feng B, LaPerle JL, Chang G, Varma MV. Renal clearance in drug discovery and development: molecular descriptors, drug transporters and disease state. Exp Opin Drug Metab Toxicol. 2010;6:939-52.
    • (2010) Exp Opin Drug Metab Toxicol , vol.6 , pp. 939-952
    • Feng, B.1    LaPerle, J.L.2    Chang, G.3    Varma, M.V.4
  • 88
    • 84865492829 scopus 로고    scopus 로고
    • In silico prediction of total human plasma clearance
    • 1:CAS:528:DC%2BC38XovVGgur8%3D 22715914
    • Berellini G, Waters NJ, Lombardo F. In silico prediction of total human plasma clearance. J Chem Inf Model. 2012;52:2069-78.
    • (2012) J Chem Inf Model , vol.52 , pp. 2069-2078
    • Berellini, G.1    Waters, N.J.2    Lombardo, F.3
  • 89
    • 84901268525 scopus 로고    scopus 로고
    • Clearance mechanism assignment and total clearance prediction in human based upon in silico models
    • Lombardo F, Obach RA, Varma MV, Stringer R, Berellini G. Clearance mechanism assignment and total clearance prediction in human based upon in silico models. J Med Chem. 2014.
    • (2014) J Med Chem.
    • Lombardo, F.1    Obach, R.A.2    Varma, M.V.3    Stringer, R.4    Berellini, G.5
  • 90
    • 77954917978 scopus 로고    scopus 로고
    • In silico classification of major clearance pathways of drugs with their physiochemical parameters
    • 1:CAS:528:DC%2BC3cXhtVWktrnM 20423955
    • Kusama M, Toshimoto K, Maeda K, Hirai Y, Imai S, Chiba K, et al. In silico classification of major clearance pathways of drugs with their physiochemical parameters. Drug Metab Dispos. 2010;38:1362-70.
    • (2010) Drug Metab Dispos , vol.38 , pp. 1362-1370
    • Kusama, M.1    Toshimoto, K.2    Maeda, K.3    Hirai, Y.4    Imai, S.5    Chiba, K.6
  • 91
    • 84907180162 scopus 로고    scopus 로고
    • Quantitative prediction of transporter- and enzyme-mediated clinical drug-drug interactions of organic anion-transporting polypeptide 1B1 substrates using a mechanistic net-effect model
    • 25107633
    • Varma MV, Bi YA, Kimoto E, Lin J. Quantitative prediction of transporter- and enzyme-mediated clinical drug-drug interactions of organic anion-transporting polypeptide 1B1 substrates using a mechanistic net-effect model. J Pharmacol Exp Ther. 2014;351:214-23.
    • (2014) J Pharmacol Exp Ther , vol.351 , pp. 214-223
    • Varma, M.V.1    Bi, Y.A.2    Kimoto, E.3    Lin, J.4
  • 92
    • 79953901458 scopus 로고    scopus 로고
    • Organic anion transporting polypeptide 1B1: A genetically polymorphic transporter of major importance for hepatic drug uptake
    • 1:CAS:528:DC%2BC3MXjslGis7w%3D 21245207
    • Niemi M, Pasanen MK, Neuvonen PJ. Organic anion transporting polypeptide 1B1: a genetically polymorphic transporter of major importance for hepatic drug uptake. Pharmacol Rev. 2011;63:157-81.
    • (2011) Pharmacol Rev , vol.63 , pp. 157-181
    • Niemi, M.1    Pasanen, M.K.2    Neuvonen, P.J.3
  • 93
    • 78049385182 scopus 로고    scopus 로고
    • Using open source computational tools for predicting human metabolic stability and additional absorption, distribution, metabolism, excretion, and toxicity properties
    • 1:CAS:528:DC%2BC3cXhsVWqurjN 20693417
    • Gupta RR, Gifford EM, Liston T, Waller CL, Hohman M, Bunin BA, et al. Using open source computational tools for predicting human metabolic stability and additional absorption, distribution, metabolism, excretion, and toxicity properties. Drug Metab Dispos. 2010;38:2083-90.
    • (2010) Drug Metab Dispos , vol.38 , pp. 2083-2090
    • Gupta, R.R.1    Gifford, E.M.2    Liston, T.3    Waller, C.L.4    Hohman, M.5    Bunin, B.A.6
  • 94
    • 33751171573 scopus 로고    scopus 로고
    • Antiplatelet agents aspirin and clopidogrel are hydrolyzed by distinct carboxylesterases, and clopidogrel is transesterificated in the presence of ethyl alcohol
    • 1:CAS:528:DC%2BD28Xht1yltLvP 16943252
    • Tang M, Mukundan M, Yang J, Charpentier N, LeCluyse EL, Black C, et al. Antiplatelet agents aspirin and clopidogrel are hydrolyzed by distinct carboxylesterases, and clopidogrel is transesterificated in the presence of ethyl alcohol. J Pharmacol Exp Ther. 2006;319:1467-76.
    • (2006) J Pharmacol Exp Ther , vol.319 , pp. 1467-1476
    • Tang, M.1    Mukundan, M.2    Yang, J.3    Charpentier, N.4    LeCluyse, E.L.5    Black, C.6
  • 95
    • 0030766633 scopus 로고    scopus 로고
    • Clinical pharmacokinetics of diclofenac. Therapeutic insights and pitfalls
    • 1:CAS:528:DyaK2sXmsVeltr4%3D 9314611
    • Davies NM, Anderson KE. Clinical pharmacokinetics of diclofenac. Therapeutic insights and pitfalls. Clin Pharmacokinet. 1997;33:184-213.
    • (1997) Clin Pharmacokinet , vol.33 , pp. 184-213
    • Davies, N.M.1    Anderson, K.E.2
  • 96
    • 0035038561 scopus 로고    scopus 로고
    • Characterization of rat and human UDP-glucuronosyltransferases responsible for the in vitro glucuronidation of diclofenac
    • 1:CAS:528:DC%2BD3MXjtV2rs74%3D
    • King C, Tang W, Ngui J, Tephly T, Braun M. Characterization of rat and human UDP-glucuronosyltransferases responsible for the in vitro glucuronidation of diclofenac. Toxicol Sci Off J Soc Toxicol. 2001;61:49-53.
    • (2001) Toxicol Sci off J Soc Toxicol , vol.61 , pp. 49-53
    • King, C.1    Tang, W.2    Ngui, J.3    Tephly, T.4    Braun, M.5
  • 97
    • 3042641177 scopus 로고    scopus 로고
    • An evaluation of the in vitro metabolism data for predicting the clearance and drug-drug interaction potential of CYP2C9 substrates
    • 1:CAS:528:DC%2BD2cXltlWqt7c%3D
    • Andersson TB, Bredberg E, Ericsson H, Sjoberg H. An evaluation of the in vitro metabolism data for predicting the clearance and drug-drug interaction potential of CYP2C9 substrates. Drug Metab Dis Biol Fate Chem. 2004;32:715-21.
    • (2004) Drug Metab Dis Biol Fate Chem , vol.32 , pp. 715-721
    • Andersson, T.B.1    Bredberg, E.2    Ericsson, H.3    Sjoberg, H.4
  • 98
    • 0033790094 scopus 로고    scopus 로고
    • Automated definition of the enzymology of drug oxidation by the major human drug metabolizing cytochrome P450s
    • 1:CAS:528:DC%2BD3cXnsFOlsb4%3D
    • McGinnity DF, Parker AJ, Soars M, Riley RJ. Automated definition of the enzymology of drug oxidation by the major human drug metabolizing cytochrome P450s. Drug Metab Dis Biol Fate Chem. 2000;28:1327-34.
    • (2000) Drug Metab Dis Biol Fate Chem , vol.28 , pp. 1327-1334
    • McGinnity, D.F.1    Parker, A.J.2    Soars, M.3    Riley, R.J.4
  • 100
    • 84866619971 scopus 로고    scopus 로고
    • Influence of UDP-glucuronosyltransferase polymorphisms on valproic acid pharmacokinetics in Chinese epilepsy patients
    • 1:CAS:528:DC%2BC38XhtlGhu7rJ 22476391
    • Chu XM, Zhang LF, Wang GJ, Zhang SN, Zhou JH, Hao HP. Influence of UDP-glucuronosyltransferase polymorphisms on valproic acid pharmacokinetics in Chinese epilepsy patients. Eur J Clin Pharmacol. 2012;68:1395-401.
    • (2012) Eur J Clin Pharmacol , vol.68 , pp. 1395-1401
    • Chu, X.M.1    Zhang, L.F.2    Wang, G.J.3    Zhang, S.N.4    Zhou, J.H.5    Hao, H.P.6
  • 101
    • 0034872136 scopus 로고    scopus 로고
    • Pharmacogenetics of warfarin elimination and its clinical implications
    • 1:CAS:528:DC%2BD3MXmsl2gsb4%3D 11523725
    • Takahashi H, Echizen H. Pharmacogenetics of warfarin elimination and its clinical implications. Clin Pharmacokinet. 2001;40:587-603.
    • (2001) Clin Pharmacokinet , vol.40 , pp. 587-603
    • Takahashi, H.1    Echizen, H.2
  • 102
    • 0242383181 scopus 로고    scopus 로고
    • Clinical pharmacokinetics of atorvastatin
    • 14531725
    • Lennernas H. Clinical pharmacokinetics of atorvastatin. Clin Pharmacokinet. 2003;42:1141-60.
    • (2003) Clin Pharmacokinet , vol.42 , pp. 1141-1160
    • Lennernas, H.1
  • 103
    • 31144438802 scopus 로고    scopus 로고
    • Multiple transporters affect the disposition of atorvastatin and its two active hydroxy metabolites: Application of in vitro and ex situ systems
    • 1:CAS:528:DC%2BD28XhtlWqtrg%3D 16258024
    • Lau YY, Okochi H, Huang Y, Benet LZ. Multiple transporters affect the disposition of atorvastatin and its two active hydroxy metabolites: application of in vitro and ex situ systems. J Pharmacol Exp Ther. 2006;316:762-71.
    • (2006) J Pharmacol Exp Ther , vol.316 , pp. 762-771
    • Lau, Y.Y.1    Okochi, H.2    Huang, Y.3    Benet, L.Z.4
  • 104
    • 10944231356 scopus 로고    scopus 로고
    • Clinical pharmacology of bosentan, a dual endothelin receptor antagonist
    • 1:CAS:528:DC%2BD2MXnvFShuw%3D%3D 15568889
    • Dingemanse J, van Giersbergen PL. Clinical pharmacology of bosentan, a dual endothelin receptor antagonist. Clin Pharmacokinet. 2004;43:1089-115.
    • (2004) Clin Pharmacokinet , vol.43 , pp. 1089-1115
    • Dingemanse, J.1    Van Giersbergen, P.L.2
  • 105
    • 4644301418 scopus 로고    scopus 로고
    • Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: Analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil
    • 1:CAS:528:DC%2BD2cXosVKrt7g%3D 15194707
    • Shitara Y, Hirano M, Sato H, Sugiyama Y. Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. J Pharmacol Exp Ther. 2004;311:228-36.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 228-236
    • Shitara, Y.1    Hirano, M.2    Sato, H.3    Sugiyama, Y.4
  • 106
    • 33845420011 scopus 로고    scopus 로고
    • Drug interactions with lipid-lowering drugs: Mechanisms and clinical relevance
    • 1:CAS:528:DC%2BD28XhtleisLfE 17178259
    • Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid-lowering drugs: mechanisms and clinical relevance. Clin Pharmacol Ther. 2006;80:565-81.
    • (2006) Clin Pharmacol Ther , vol.80 , pp. 565-581
    • Neuvonen, P.J.1    Niemi, M.2    Backman, J.T.3
  • 107
    • 4644364576 scopus 로고    scopus 로고
    • Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans
    • 1:CAS:528:DC%2BD2cXosVKrtrg%3D 15159445
    • Hirano M, Maeda K, Shitara Y, Sugiyama Y. Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J Pharmacol Exp Ther. 2004;311:139-46.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 139-146
    • Hirano, M.1    Maeda, K.2    Shitara, Y.3    Sugiyama, Y.4
  • 108
    • 3242695305 scopus 로고    scopus 로고
    • Effect of gemfibrozil on the metabolism of pitavastatin - Determining the best animal model for human CYP and UGT activities
    • 1:CAS:528:DC%2BD2cXms1eitbw%3D 15283301
    • Fujino H, Saito T, Tsunenari Y, Kojima J. Effect of gemfibrozil on the metabolism of pitavastatin - determining the best animal model for human CYP and UGT activities. Drug Metabol Drug Interact. 2004;20:25-42.
    • (2004) Drug Metabol Drug Interact , vol.20 , pp. 25-42
    • Fujino, H.1    Saito, T.2    Tsunenari, Y.3    Kojima, J.4
  • 109
    • 84907588697 scopus 로고    scopus 로고
    • Physiologically based pharmacokinetic prediction of telmisartan in human
    • 1:CAS:528:DC%2BC2MXmsVSmuro%3D
    • Li R, Ghosh A, Maurer TS, Kimoto E, Barton HA. Physiologically based pharmacokinetic prediction of telmisartan in human. Drug Metab Dis Biol Fate Chem. 2014;42:1646-55.
    • (2014) Drug Metab Dis Biol Fate Chem , vol.42 , pp. 1646-1655
    • Li, R.1    Ghosh, A.2    Maurer, T.S.3    Kimoto, E.4    Barton, H.A.5
  • 110
    • 0034045094 scopus 로고    scopus 로고
    • Pharmacokinetic interaction of abacavir (1592U89) and ethanol in human immunodeficiency virus-infected adults
    • 89933 1:CAS:528:DC%2BD3cXjs1Ols7g%3D 10817729
    • McDowell JA, Chittick GE, Stevens CP, Edwards KD, Stein DS. Pharmacokinetic interaction of abacavir (1592U89) and ethanol in human immunodeficiency virus-infected adults. Antimicrob Agents Chemother. 2000;44:1686-90.
    • (2000) Antimicrob Agents Chemother , vol.44 , pp. 1686-1690
    • McDowell, J.A.1    Chittick, G.E.2    Stevens, C.P.3    Edwards, K.D.4    Stein, D.S.5
  • 111
    • 0035187828 scopus 로고    scopus 로고
    • Pharmacokinetic aspects of treating infections in the intensive care unit: Focus on drug interactions
    • 1:CAS:528:DC%2BD3MXpt1ejt7s%3D 11735605
    • Pea F, Furlanut M. Pharmacokinetic aspects of treating infections in the intensive care unit: focus on drug interactions. Clin Pharmacokinet. 2001;40:833-68.
    • (2001) Clin Pharmacokinet , vol.40 , pp. 833-868
    • Pea, F.1    Furlanut, M.2
  • 112
    • 84895520896 scopus 로고    scopus 로고
    • Physiologically based pharmacokinetic modeling of CYP3A4 induction by rifampicin in human: Influence of time between substrate and inducer administration
    • 1:CAS:528:DC%2BC2cXmt1GlurY%3D 24530864
    • Baneyx G, Parrott N, Meille C, Iliadis A, Lave T. Physiologically based pharmacokinetic modeling of CYP3A4 induction by rifampicin in human: influence of time between substrate and inducer administration. Eur J Pharm Sci. 2014;56:1-15.
    • (2014) Eur J Pharm Sci , vol.56 , pp. 1-15
    • Baneyx, G.1    Parrott, N.2    Meille, C.3    Iliadis, A.4    Lave, T.5
  • 114
    • 7244224993 scopus 로고    scopus 로고
    • Interactions between grapefruit juice and cardiovascular drugs
    • 1:CAS:528:DC%2BD2cXhtVShtLjN
    • Bailey DG, Dresser GK. Interactions between grapefruit juice and cardiovascular drugs. Am J Cardiovasc Drugs Drugs Devices Interv. 2004;4:281-97.
    • (2004) Am J Cardiovasc Drugs Drugs Devices Interv , vol.4 , pp. 281-297
    • Bailey, D.G.1    Dresser, G.K.2
  • 115
    • 6044261115 scopus 로고    scopus 로고
    • Pharmacokinetics of budesonide (Entocort EC) capsules for Crohn's disease
    • 15355126
    • Edsbacker S, Andersson T. Pharmacokinetics of budesonide (Entocort EC) capsules for Crohn's disease. Clin Pharmacokinet. 2004;43:803-21.
    • (2004) Clin Pharmacokinet , vol.43 , pp. 803-821
    • Edsbacker, S.1    Andersson, T.2
  • 116
    • 0029145903 scopus 로고
    • Drug interactions and the cytochrome P450 system. The role of cytochrome P450 1A2
    • 8846619
    • Brosen K. Drug interactions and the cytochrome P450 system. The role of cytochrome P450 1A2. Clin Pharmacokinet. 1995;29(1):20-5.
    • (1995) Clin Pharmacokinet , vol.29 , Issue.1 , pp. 20-25
    • Brosen, K.1
  • 117
    • 24944464940 scopus 로고    scopus 로고
    • Contribution of human hepatic cytochrome p450 isoforms to the metabolism of psychotropic drugs
    • 1:CAS:528:DC%2BD2MXhtFWjurvP 16141545
    • Niwa T, Shiraga T, Ishii I, Kagayama A, Takagi A. Contribution of human hepatic cytochrome p450 isoforms to the metabolism of psychotropic drugs. Biol Pharm Bull. 2005;28:1711-6.
    • (2005) Biol Pharm Bull , vol.28 , pp. 1711-1716
    • Niwa, T.1    Shiraga, T.2    Ishii, I.3    Kagayama, A.4    Takagi, A.5
  • 118
    • 0030875423 scopus 로고    scopus 로고
    • Role of CYP3A4 in human hepatic diltiazem N-demethylation: Inhibition of CYP3A4 activity by oxidized diltiazem metabolites
    • 1:CAS:528:DyaK2sXksFyitb0%3D
    • Sutton D, Butler AM, Nadin L, Murray M. Role of CYP3A4 in human hepatic diltiazem N-demethylation: inhibition of CYP3A4 activity by oxidized diltiazem metabolites. J Pharm Exp Ther. 1997;282:294-300.
    • (1997) J Pharm Exp Ther , vol.282 , pp. 294-300
    • Sutton, D.1    Butler, A.M.2    Nadin, L.3    Murray, M.4
  • 119
    • 70449704111 scopus 로고    scopus 로고
    • Clinical pharmacokinetics of tyrosine kinase inhibitors
    • 19733976
    • van Erp NP, Gelderblom H, Guchelaar HJ. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev. 2009;35:692-706.
    • (2009) Cancer Treat Rev , vol.35 , pp. 692-706
    • Van Erp, N.P.1    Gelderblom, H.2    Guchelaar, H.J.3
  • 120
    • 34547641389 scopus 로고    scopus 로고
    • General framework for the quantitative prediction of CYP3A4-mediated oral drug interactions based on the AUC increase by coadministration of standard drugs
    • 1:CAS:528:DC%2BD2sXhtVKgs7%2FL 17655375
    • Ohno Y, Hisaka A, Suzuki H. General framework for the quantitative prediction of CYP3A4-mediated oral drug interactions based on the AUC increase by coadministration of standard drugs. Clin Pharmacokinet. 2007;46:681-96.
    • (2007) Clin Pharmacokinet , vol.46 , pp. 681-696
    • Ohno, Y.1    Hisaka, A.2    Suzuki, H.3
  • 122
    • 37549060373 scopus 로고    scopus 로고
    • Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination
    • 3691104 1:CAS:528:DC%2BD2sXhsVOrtrbF 18098064
    • Jeong H, Choi S, Song JW, Chen H, Fischer JH. Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Xenobiotica. 2008;38:62-75.
    • (2008) Xenobiotica , vol.38 , pp. 62-75
    • Jeong, H.1    Choi, S.2    Song, J.W.3    Chen, H.4    Fischer, J.H.5
  • 124
    • 0037023533 scopus 로고    scopus 로고
    • The influence of P-glycoprotein on morphine transport in Caco-2 cells. Comparison with paclitaxel
    • 1:CAS:528:DC%2BD38XivVSjur0%3D 11959083
    • Crowe A. The influence of P-glycoprotein on morphine transport in Caco-2 cells. Comparison with paclitaxel. Eur J Pharmacol. 2002;440:7-16.
    • (2002) Eur J Pharmacol , vol.440 , pp. 7-16
    • Crowe, A.1
  • 125
    • 84896446184 scopus 로고    scopus 로고
    • Morphine glucuronidation and glucosidation represent complementary metabolic pathways that are both catalyzed by UDP-glucuronosyltransferase 2B7: Kinetic, inhibition, and molecular modeling studies
    • 24459244
    • Chau N, Elliot DJ, Lewis BC, Burns K, Johnston MR, Mackenzie PI, et al. Morphine glucuronidation and glucosidation represent complementary metabolic pathways that are both catalyzed by UDP-glucuronosyltransferase 2B7: kinetic, inhibition, and molecular modeling studies. J Pharmacol Exp Ther. 2014;349:126-37.
    • (2014) J Pharmacol Exp Ther , vol.349 , pp. 126-137
    • Chau, N.1    Elliot, D.J.2    Lewis, B.C.3    Burns, K.4    Johnston, M.R.5    MacKenzie, P.I.6
  • 126
    • 77952302682 scopus 로고    scopus 로고
    • Selection of alternative CYP3A4 probe substrates for clinical drug interaction studies using in vitro data and in vivo simulation
    • 1:CAS:528:DC%2BC3cXmvVCjtL8%3D
    • Foti RS, Rock DA, Wienkers LC, Wahlstrom JL. Selection of alternative CYP3A4 probe substrates for clinical drug interaction studies using in vitro data and in vivo simulation. Drug Metab Dis Biol Fate Chem. 2010;38:981-7.
    • (2010) Drug Metab Dis Biol Fate Chem , vol.38 , pp. 981-987
    • Foti, R.S.1    Rock, D.A.2    Wienkers, L.C.3    Wahlstrom, J.L.4
  • 127
    • 0037437688 scopus 로고    scopus 로고
    • Stereospecific analysis of omeprazole in human plasma as a probe for CYP2C19 phenotype
    • 1:CAS:528:DC%2BD38XpsFWiu70%3D 12485723
    • Kanazawa H, Okada A, Higaki M, Yokota H, Mashige F, Nakahara K. Stereospecific analysis of omeprazole in human plasma as a probe for CYP2C19 phenotype. J Pharm Biomed Anal. 2003;30:1817-24.
    • (2003) J Pharm Biomed Anal , vol.30 , pp. 1817-1824
    • Kanazawa, H.1    Okada, A.2    Higaki, M.3    Yokota, H.4    Mashige, F.5    Nakahara, K.6
  • 128
    • 79551533310 scopus 로고    scopus 로고
    • Regio- and stereoselective oxidation of propranolol enantiomers by human CYP2D6, cynomolgus monkey CYP2D17 and marmoset CYP2D19
    • 1:CAS:528:DC%2BC3MXhtlGktLo%3D 21184751
    • Narimatsu S, Nakata T, Shimizudani T, Nagaoka K, Nakura H, Masuda K, et al. Regio- and stereoselective oxidation of propranolol enantiomers by human CYP2D6, cynomolgus monkey CYP2D17 and marmoset CYP2D19. Chem Biol Interact. 2011;189:146-52.
    • (2011) Chem Biol Interact , vol.189 , pp. 146-152
    • Narimatsu, S.1    Nakata, T.2    Shimizudani, T.3    Nagaoka, K.4    Nakura, H.5    Masuda, K.6
  • 129
    • 23944512775 scopus 로고    scopus 로고
    • The clinical pharmacokinetics of phosphodiesterase-5 inhibitors for erectile dysfunction
    • 1:CAS:528:DC%2BD2MXhtVemsrjE 16100293
    • Gupta M, Kovar A, Meibohm B. The clinical pharmacokinetics of phosphodiesterase-5 inhibitors for erectile dysfunction. J Clin Pharmacol. 2005;45:987-1003.
    • (2005) J Clin Pharmacol , vol.45 , pp. 987-1003
    • Gupta, M.1    Kovar, A.2    Meibohm, B.3
  • 132
    • 0038722339 scopus 로고    scopus 로고
    • Ziprasidone metabolism, aldehyde oxidase, and clinical implications
    • 1:CAS:528:DC%2BD3sXltFKjt7c%3D 12826984
    • Beedham C, Miceli JJ, Obach RS. Ziprasidone metabolism, aldehyde oxidase, and clinical implications. J Clin Psychopharmacol. 2003;23:229-32.
    • (2003) J Clin Psychopharmacol , vol.23 , pp. 229-232
    • Beedham, C.1    Miceli, J.J.2    Obach, R.S.3
  • 133
    • 33645101081 scopus 로고    scopus 로고
    • Interactions of amoxicillin and cefaclor with human renal organic anion and peptide transporters
    • 1:CAS:528:DC%2BD28XjtlKns7k%3D
    • Li M, Anderson GD, Phillips BR, Kong W, Shen DD, Wang J. Interactions of amoxicillin and cefaclor with human renal organic anion and peptide transporters. Drug Metab Dis Biol Fate Chem. 2006;34:547-55.
    • (2006) Drug Metab Dis Biol Fate Chem , vol.34 , pp. 547-555
    • Li, M.1    Anderson, G.D.2    Phillips, B.R.3    Kong, W.4    Shen, D.D.5    Wang, J.6
  • 134
    • 24344461838 scopus 로고    scopus 로고
    • Human organic anion transporter hOAT3 is a potent transporter of cephalosporin antibiotics, in comparison with hOAT1
    • 1:CAS:528:DC%2BD2MXpvFegsrc%3D 16098483
    • Ueo H, Motohashi H, Katsura T, Inui K. Human organic anion transporter hOAT3 is a potent transporter of cephalosporin antibiotics, in comparison with hOAT1. Biochem Pharmacol. 2005;70:1104-13.
    • (2005) Biochem Pharmacol , vol.70 , pp. 1104-1113
    • Ueo, H.1    Motohashi, H.2    Katsura, T.3    Inui, K.4
  • 135
    • 10744233574 scopus 로고    scopus 로고
    • Interactions of human organic anion transporters with diuretics
    • 1:CAS:528:DC%2BD2cXhvVWntbk%3D
    • Hasannejad H, Takeda M, Taki K, Shin HJ, Babu E, Jutabha P, et al. Interactions of human organic anion transporters with diuretics. J Pharm Exp Ther. 2004;308:1021-9.
    • (2004) J Pharm Exp Ther , vol.308 , pp. 1021-1029
    • Hasannejad, H.1    Takeda, M.2    Taki, K.3    Shin, H.J.4    Babu, E.5    Jutabha, P.6
  • 136
    • 84879336491 scopus 로고    scopus 로고
    • Benzylpenicillin inhibits the renal excretion of acyclovir by OAT1 and OAT3
    • 1:CAS:528:DC%2BC3sXhsFSmurnO 23744435
    • Ye J, Liu Q, Wang C, Meng Q, Sun H, Peng J, et al. Benzylpenicillin inhibits the renal excretion of acyclovir by OAT1 and OAT3. Pharmacol Rep PR. 2013;65:505-12.
    • (2013) Pharmacol Rep PR , vol.65 , pp. 505-512
    • Ye, J.1    Liu, Q.2    Wang, C.3    Meng, Q.4    Sun, H.5    Peng, J.6
  • 137
    • 84902287467 scopus 로고    scopus 로고
    • MDR1 and OAT1/OAT3 mediate the drug-drug interaction between puerarin and methotrexate
    • 1:CAS:528:DC%2BC3sXhslymu7bO 24242937
    • Liu Q, Wang C, Meng Q, Huo X, Sun H, Peng J, et al. MDR1 and OAT1/OAT3 mediate the drug-drug interaction between puerarin and methotrexate. Pharm Res. 2014;31:1120-32.
    • (2014) Pharm Res , vol.31 , pp. 1120-1132
    • Liu, Q.1    Wang, C.2    Meng, Q.3    Huo, X.4    Sun, H.5    Peng, J.6
  • 138
    • 84887992470 scopus 로고    scopus 로고
    • Interaction of immunosuppressive drugs with human organic anion transporter (OAT) 1 and OAT3, and multidrug resistance-associated protein (MRP) 2 and MRP4
    • 1:CAS:528:DC%2BC3sXhsFeksrnL
    • El-Sheikh AA, Greupink R, Wortelboer HM, van den Heuvel JJ, Schreurs M, Koenderink JB, et al. Interaction of immunosuppressive drugs with human organic anion transporter (OAT) 1 and OAT3, and multidrug resistance-associated protein (MRP) 2 and MRP4. Transl Re J Lab Clin Med. 2013;162:398-409.
    • (2013) Transl Re J Lab Clin Med , vol.162 , pp. 398-409
    • El-Sheikh, A.A.1    Greupink, R.2    Wortelboer, H.M.3    Van Den Heuvel, J.J.4    Schreurs, M.5    Koenderink, J.B.6
  • 139
    • 52949104006 scopus 로고    scopus 로고
    • Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin
    • 1:CAS:528:DC%2BD1cXhtF2jtLbJ
    • Kitamura S, Maeda K, Wang Y, Sugiyama Y. Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin. Drug Metab Dis Biol Fate Chem. 2008;36:2014-23.
    • (2008) Drug Metab Dis Biol Fate Chem , vol.36 , pp. 2014-2023
    • Kitamura, S.1    Maeda, K.2    Wang, Y.3    Sugiyama, Y.4
  • 140
    • 84864400498 scopus 로고    scopus 로고
    • Inhibitory effect of JBP485 on renal excretion of acyclovir by the inhibition of OAT1 and OAT3
    • 1:CAS:528:DC%2BC38Xht1Kku7bE 22728397
    • Ye J, Liu Q, Wang C, Meng Q, Peng J, Sun H, et al. Inhibitory effect of JBP485 on renal excretion of acyclovir by the inhibition of OAT1 and OAT3. Eur J Pharm Sci. 2012;47:341-6.
    • (2012) Eur J Pharm Sci , vol.47 , pp. 341-346
    • Ye, J.1    Liu, Q.2    Wang, C.3    Meng, Q.4    Peng, J.5    Sun, H.6
  • 141
    • 25644452793 scopus 로고    scopus 로고
    • A species difference in the transport activities of H2 receptor antagonists by rat and human renal organic anion and cation transporters
    • 1:CAS:528:DC%2BD2MXhtVOks7vF 16006492
    • Tahara H, Kusuhara H, Endou H, Koepsell H, Imaoka T, Fuse E, et al. A species difference in the transport activities of H2 receptor antagonists by rat and human renal organic anion and cation transporters. J Pharmacol Exp Ther. 2005;315:337-45.
    • (2005) J Pharmacol Exp Ther , vol.315 , pp. 337-345
    • Tahara, H.1    Kusuhara, H.2    Endou, H.3    Koepsell, H.4    Imaoka, T.5    Fuse, E.6
  • 142
    • 84885181188 scopus 로고    scopus 로고
    • Role of organic cation transporter OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2-K for transport and drug interactions of the antiviral lamivudine
    • 23876341
    • Muller F, Konig J, Hoier E, Mandery K, Fromm MF. Role of organic cation transporter OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2-K for transport and drug interactions of the antiviral lamivudine. Biochem Pharmacol. 2013;86:808-15.
    • (2013) Biochem Pharmacol , vol.86 , pp. 808-815
    • Muller, F.1    Konig, J.2    Hoier, E.3    Mandery, K.4    Fromm, M.F.5
  • 143
    • 84878889895 scopus 로고    scopus 로고
    • Effects of OCT2 c.602C > T genetic variant on the pharmacokinetics of lamivudine
    • 1:CAS:528:DC%2BC3sXos1yns78%3D 23252721
    • Choi CI, Bae JW, Keum SK, Lee YJ, Lee HI, Jang CG, et al. Effects of OCT2 c.602C > T genetic variant on the pharmacokinetics of lamivudine. Xenobiotica. 2013;43:636-40.
    • (2013) Xenobiotica , vol.43 , pp. 636-640
    • Choi, C.I.1    Bae, J.W.2    Keum, S.K.3    Lee, Y.J.4    Lee, H.I.5    Jang, C.G.6
  • 144
    • 27744570434 scopus 로고    scopus 로고
    • Differential substrate and inhibitory activities of ranitidine and famotidine toward human organic cation transporter 1 (hOCT1; SLC22A1), hOCT2 (SLC22A2), and hOCT3 (SLC22A3)
    • 1:CAS:528:DC%2BD2MXhtlalurnE 16141367
    • Bourdet DL, Pritchard JB, Thakker DR. Differential substrate and inhibitory activities of ranitidine and famotidine toward human organic cation transporter 1 (hOCT1; SLC22A1), hOCT2 (SLC22A2), and hOCT3 (SLC22A3). J Pharmacol Exp Ther. 2005;315:1288-97.
    • (2005) J Pharmacol Exp Ther , vol.315 , pp. 1288-1297
    • Bourdet, D.L.1    Pritchard, J.B.2    Thakker, D.R.3
  • 145
    • 34247236764 scopus 로고    scopus 로고
    • Transport of the dipeptidyl peptidase-4 inhibitor sitagliptin by human organic anion transporter 3, organic anion transporting polypeptide 4C1, and multidrug resistance P-glycoprotein
    • 1:CAS:528:DC%2BD2sXltFehurk%3D 17314201
    • Chu XY, Bleasby K, Yabut J, Cai X, Chan GH, Hafey MJ, et al. Transport of the dipeptidyl peptidase-4 inhibitor sitagliptin by human organic anion transporter 3, organic anion transporting polypeptide 4C1, and multidrug resistance P-glycoprotein. J Pharmacol Exp Ther. 2007;321:673-83.
    • (2007) J Pharmacol Exp Ther , vol.321 , pp. 673-683
    • Chu, X.Y.1    Bleasby, K.2    Yabut, J.3    Cai, X.4    Chan, G.H.5    Hafey, M.J.6
  • 146
    • 0034525310 scopus 로고    scopus 로고
    • Modelling human cytochromes P450 for evaluating drug metabolism: An update
    • 1:CAS:528:DC%2BD3MXis1Ggsw%3D%3D 11201309
    • Lewis DF. Modelling human cytochromes P450 for evaluating drug metabolism: an update. Drug Metabol Drug Interact. 2000;16:307-24.
    • (2000) Drug Metabol Drug Interact , vol.16 , pp. 307-324
    • Lewis, D.F.1
  • 147
    • 65449166750 scopus 로고    scopus 로고
    • Prediction of human pharmacokinetics from preclinical information: Comparative accuracy of quantitative prediction approaches
    • 1:CAS:528:DC%2BD1MXlsFWmu74%3D 19299532
    • Hosea NA, Collard WT, Cole S, Maurer TS, Fang RX, Jones H, et al. Prediction of human pharmacokinetics from preclinical information: comparative accuracy of quantitative prediction approaches. J Clin Pharmacol. 2009;49:513-33.
    • (2009) J Clin Pharmacol , vol.49 , pp. 513-533
    • Hosea, N.A.1    Collard, W.T.2    Cole, S.3    Maurer, T.S.4    Fang, R.X.5    Jones, H.6
  • 148
    • 67349229228 scopus 로고    scopus 로고
    • Rifampicin alters atorvastatin plasma concentration on the basis of SLCO1B1 521T>C polymorphism
    • 1:CAS:528:DC%2BD1MXms1Kls7c%3D
    • He YJ, Zhang W, Chen Y, Guo D, Tu JH, Xu LY, et al. Rifampicin alters atorvastatin plasma concentration on the basis of SLCO1B1 521T>C polymorphism. Clin Chim Acta Int J Clin Chem. 2009;405:49-52.
    • (2009) Clin Chim Acta Int J Clin Chem , vol.405 , pp. 49-52
    • He, Y.J.1    Zhang, W.2    Chen, Y.3    Guo, D.4    Tu, J.H.5    Xu, L.Y.6
  • 150
    • 0033745108 scopus 로고    scopus 로고
    • Itraconazole alters the pharmacokinetics of atorvastatin to a greater extent than either cerivastatin or pravastatin
    • 1:CAS:528:DC%2BD3cXot1Wjtr8%3D 11061579
    • Mazzu AL, Lasseter KC, Shamblen EC, Agarwal V, Lettieri J, Sundaresen P. Itraconazole alters the pharmacokinetics of atorvastatin to a greater extent than either cerivastatin or pravastatin. Clin Pharmacol Ther. 2000;68:391-400.
    • (2000) Clin Pharmacol Ther , vol.68 , pp. 391-400
    • Mazzu, A.L.1    Lasseter, K.C.2    Shamblen, E.C.3    Agarwal, V.4    Lettieri, J.5    Sundaresen, P.6
  • 151
    • 0036209898 scopus 로고    scopus 로고
    • A study of the interaction potential of azithromycin and clarithromycin with atorvastatin in healthy volunteers
    • 1:CAS:528:DC%2BD38Xjt1ynsrg%3D 11936570
    • Amsden GW, Kuye O, Wei GC. A study of the interaction potential of azithromycin and clarithromycin with atorvastatin in healthy volunteers. J Clin Pharmacol. 2002;42:444-9.
    • (2002) J Clin Pharmacol , vol.42 , pp. 444-449
    • Amsden, G.W.1    Kuye, O.2    Wei, G.C.3
  • 152
    • 0032806143 scopus 로고    scopus 로고
    • Erythromycin coadministration increases plasma atorvastatin concentrations
    • 1:CAS:528:DC%2BD3cXivFajsQ%3D%3D 10234598
    • Siedlik PH, Olson SC, Yang BB, Stern RH. Erythromycin coadministration increases plasma atorvastatin concentrations. J Clin Pharmacol. 1999;39:501-4.
    • (1999) J Clin Pharmacol , vol.39 , pp. 501-504
    • Siedlik, P.H.1    Olson, S.C.2    Yang, B.B.3    Stern, R.H.4
  • 153
    • 84906508843 scopus 로고    scopus 로고
    • Pitavastatin is a more sensitive and selective organic anion-transporting polypeptide 1B clinical probe than rosuvastatin
    • 4243909 1:CAS:528:DC%2BC2cXhsVWltbnJ 24617605
    • Prueksaritanont T, Chu X, Evers R, Klopfer SO, Caro L, Kothare PA, et al. Pitavastatin is a more sensitive and selective organic anion-transporting polypeptide 1B clinical probe than rosuvastatin. Br J Clin Pharmacol. 2014;78:587-98.
    • (2014) Br J Clin Pharmacol , vol.78 , pp. 587-598
    • Prueksaritanont, T.1    Chu, X.2    Evers, R.3    Klopfer, S.O.4    Caro, L.5    Kothare, P.A.6
  • 154
    • 0033064071 scopus 로고    scopus 로고
    • Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporine-treated kidney transplant recipients
    • 1:CAS:528:DyaK1MXitFeqt74%3D 10096257
    • Muck W, Mai I, Fritsche L, Ochmann K, Rohde G, Unger S, et al. Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporine-treated kidney transplant recipients. Clin Pharmacol Ther. 1999;65:251-61.
    • (1999) Clin Pharmacol Ther , vol.65 , pp. 251-261
    • Muck, W.1    Mai, I.2    Fritsche, L.3    Ochmann, K.4    Rohde, G.5    Unger, S.6
  • 155
    • 0036918664 scopus 로고    scopus 로고
    • Gemfibrozil greatly increases plasma concentrations of cerivastatin
    • 1:CAS:528:DC%2BD3sXlt1SjsQ%3D%3D 12496749
    • Backman JT, Kyrklund C, Neuvonen M, Neuvonen PJ. Gemfibrozil greatly increases plasma concentrations of cerivastatin. Clin Pharmacol Ther. 2002;72:685-91.
    • (2002) Clin Pharmacol Ther , vol.72 , pp. 685-691
    • Backman, J.T.1    Kyrklund, C.2    Neuvonen, M.3    Neuvonen, P.J.4
  • 156
    • 0033920666 scopus 로고    scopus 로고
    • Effect of fluconazole on plasma fluvastatin and pravastatin concentrations
    • 1:CAS:528:DC%2BD3cXjvFCgsbo%3D 10952477
    • Kantola T, Backman JT, Niemi M, Kivisto KT, Neuvonen PJ. Effect of fluconazole on plasma fluvastatin and pravastatin concentrations. Eur J Clin Pharmacol. 2000;56:225-9.
    • (2000) Eur J Clin Pharmacol , vol.56 , pp. 225-229
    • Kantola, T.1    Backman, J.T.2    Niemi, M.3    Kivisto, K.T.4    Neuvonen, P.J.5
  • 157
    • 0035710125 scopus 로고    scopus 로고
    • Pharmacokinetics and pharmacodynamics of fluvastatin in heart transplant recipients taking cyclosporine A
    • 1:CAS:528:DC%2BD38XitVCrsb0%3D 11907637
    • Park JW, Siekmeier R, Lattke P, Merz M, Mix C, Schuler S, et al. Pharmacokinetics and pharmacodynamics of fluvastatin in heart transplant recipients taking cyclosporine A. J Cardiovasc Pharmacol Ther. 2001;6:351-61.
    • (2001) J Cardiovasc Pharmacol Ther , vol.6 , pp. 351-361
    • Park, J.W.1    Siekmeier, R.2    Lattke, P.3    Merz, M.4    Mix, C.5    Schuler, S.6
  • 158
    • 0028846142 scopus 로고
    • In vivo inhibition profile of cytochrome P450TB (CYP2C9) by (+/-)-fluvastatin
    • 1:CAS:528:DyaK28XhslKrsw%3D%3D 7586933
    • Transon C, Leemann T, Vogt N, Dayer P. in vivo inhibition profile of cytochrome P450TB (CYP2C9) by (+/-)-fluvastatin. Clin Pharmacol Ther. 1995;58:412-7.
    • (1995) Clin Pharmacol Ther , vol.58 , pp. 412-417
    • Transon, C.1    Leemann, T.2    Vogt, N.3    Dayer, P.4
  • 159
    • 0031969657 scopus 로고    scopus 로고
    • Influence of erythromycin pre- and co-treatment on single-dose pharmacokinetics of the HMG-CoA reductase inhibitor cerivastatin
    • 1:STN:280:DyaK1c3htFCgsg%3D%3D 9551706
    • Muck W, Ochmann K, Rohde G, Unger S, Kuhlmann J. Influence of erythromycin pre- and co-treatment on single-dose pharmacokinetics of the HMG-CoA reductase inhibitor cerivastatin. Eur J Clin Pharmacol. 1998;53:469-73.
    • (1998) Eur J Clin Pharmacol , vol.53 , pp. 469-473
    • Muck, W.1    Ochmann, K.2    Rohde, G.3    Unger, S.4    Kuhlmann, J.5
  • 161
    • 84859910816 scopus 로고    scopus 로고
    • Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data
    • 1:CAS:528:DC%2BC38Xmt1KnsrY%3D
    • Jones HM, Barton HA, Lai Y, Bi YA, Kimoto E, Kempshall S, et al. Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data. Drug Metab Dis Biol Fate Chem. 2012;40:1007-17.
    • (2012) Drug Metab Dis Biol Fate Chem , vol.40 , pp. 1007-1017
    • Jones, H.M.1    Barton, H.A.2    Lai, Y.3    Bi, Y.A.4    Kimoto, E.5    Kempshall, S.6
  • 162
    • 34249006024 scopus 로고    scopus 로고
    • Use of hepatocytes to assess the contribution of hepatic uptake to clearance in vivo
    • 1:CAS:528:DC%2BD2sXmt1ait7Y%3D 17344337
    • Soars MG, Grime K, Sproston JL, Webborn PJ, Riley RJ. Use of hepatocytes to assess the contribution of hepatic uptake to clearance in vivo. Drug Metab Dispos. 2007;35:859-65.
    • (2007) Drug Metab Dispos , vol.35 , pp. 859-865
    • Soars, M.G.1    Grime, K.2    Sproston, J.L.3    Webborn, P.J.4    Riley, R.J.5
  • 163
    • 84865191410 scopus 로고    scopus 로고
    • Use of mechanistic modeling to assess interindividual variability and interspecies differences in active uptake in human and rat hepatocytes
    • 3422540 22665271
    • Ménochet K, Kenworthy KE, Houston JB, Galetin A. Use of mechanistic modeling to assess interindividual variability and interspecies differences in active uptake in human and rat hepatocytes. Drug Metab Dispos. 2012;40:1744-56.
    • (2012) Drug Metab Dispos , vol.40 , pp. 1744-1756
    • Ménochet, K.1    Kenworthy, K.E.2    Houston, J.B.3    Galetin, A.4
  • 164
    • 77954362516 scopus 로고    scopus 로고
    • Sandwich-cultured hepatocytes: An in vitro model to evaluate hepatobiliary transporter-based drug interactions and hepatotoxicity
    • 3097390 1:CAS:528:DC%2BC3cXotlSmt78%3D 20109035
    • Swift B, Pfeifer ND, Brouwer KL. Sandwich-cultured hepatocytes: an in vitro model to evaluate hepatobiliary transporter-based drug interactions and hepatotoxicity. Drug Metab Rev. 2010;42:446-71.
    • (2010) Drug Metab Rev , vol.42 , pp. 446-471
    • Swift, B.1    Pfeifer, N.D.2    Brouwer, K.L.3
  • 165
    • 76149124823 scopus 로고    scopus 로고
    • Investigation of the rate-determining process in the hepatic elimination of HMG-CoA reductase inhibitors in rats and humans
    • 1:CAS:528:DC%2BC3cXhsVSnu7s%3D 19875501
    • Watanabe T, Kusuhara H, Maeda K, Kanamaru H, Saito Y, Hu Z, et al. Investigation of the rate-determining process in the hepatic elimination of HMG-CoA reductase inhibitors in rats and humans. Drug Metab Dispos. 2010;38:215-22.
    • (2010) Drug Metab Dispos , vol.38 , pp. 215-222
    • Watanabe, T.1    Kusuhara, H.2    Maeda, K.3    Kanamaru, H.4    Saito, Y.5    Hu, Z.6
  • 166
    • 70849085550 scopus 로고    scopus 로고
    • Inhibition of oral midazolam clearance by boosting doses of ritonavir, and by 4,4-dimethyl-benziso-(2H)-selenazine (ALT-2074), an experimental catalytic mimic of glutathione oxidase
    • 2810804 1:CAS:528:DC%2BC3cXkvFOqtg%3D%3D 20002087
    • Greenblatt DJ, Peters DE, Oleson LE, Harmatz JS, MacNab MW, Berkowitz N, et al. Inhibition of oral midazolam clearance by boosting doses of ritonavir, and by 4,4-dimethyl-benziso-(2H)-selenazine (ALT-2074), an experimental catalytic mimic of glutathione oxidase. Br J Clin Pharmacol. 2009;68:920-7.
    • (2009) Br J Clin Pharmacol , vol.68 , pp. 920-927
    • Greenblatt, D.J.1    Peters, D.E.2    Oleson, L.E.3    Harmatz, J.S.4    MacNab, M.W.5    Berkowitz, N.6
  • 167
    • 84879388463 scopus 로고    scopus 로고
    • Ethnic variability in the plasma exposures of OATP1B1 substrates such as HMG-CoA reductase inhibitors: A kinetic consideration of its mechanism
    • 1:CAS:528:DC%2BC3sXpvVOrur4%3D 23443754
    • Tomita Y, Maeda K, Sugiyama Y. Ethnic variability in the plasma exposures of OATP1B1 substrates such as HMG-CoA reductase inhibitors: a kinetic consideration of its mechanism. Clin Pharmacol Ther. 2013;94:37-51.
    • (2013) Clin Pharmacol Ther , vol.94 , pp. 37-51
    • Tomita, Y.1    Maeda, K.2    Sugiyama, Y.3
  • 168
    • 84875161979 scopus 로고    scopus 로고
    • Transporter biology in drug approval: Regulatory aspects
    • 1:CAS:528:DC%2BC3sXksVylu7g%3D
    • Maeda K, Sugiyama Y. Transporter biology in drug approval: regulatory aspects. Mol Asp Med. 2013;34:711-8.
    • (2013) Mol Asp Med , vol.34 , pp. 711-718
    • Maeda, K.1    Sugiyama, Y.2
  • 169
    • 79956123094 scopus 로고    scopus 로고
    • Prediction of human renal clearance from preclinical species for a diverse set of drugs that exhibit both active secretion and net reabsorption
    • 1:CAS:528:DC%2BC3MXmvFGnsrY%3D 21357702
    • Paine SW, Ménochet K, Denton R, McGinnity DF, Riley RJ. Prediction of human renal clearance from preclinical species for a diverse set of drugs that exhibit both active secretion and net reabsorption. Drug Metab Dispos. 2011;39:1008-13.
    • (2011) Drug Metab Dispos , vol.39 , pp. 1008-1013
    • Paine, S.W.1    Ménochet, K.2    Denton, R.3    McGinnity, D.F.4    Riley, R.J.5
  • 170
    • 84904904807 scopus 로고    scopus 로고
    • In vitro and in vivo approaches to characterize transporter-mediated disposition in drug discovery
    • 1:CAS:528:DC%2BC2cXht1alu7vJ
    • Feng B, Varma MV, Costales C, Zhang H, Tremaine L. In vitro and in vivo approaches to characterize transporter-mediated disposition in drug discovery. Exp Opin Drug Disc. 2014;9:873-90.
    • (2014) Exp Opin Drug Disc , vol.9 , pp. 873-890
    • Feng, B.1    Varma, M.V.2    Costales, C.3    Zhang, H.4    Tremaine, L.5
  • 171
    • 0028948839 scopus 로고
    • A theoretical basis for a biopharmaceutic drug classification: The correlation of in vitro drug product dissolution and in vivo bioavailability
    • 1:CAS:528:DyaK2MXksVGqur8%3D 7617530
    • Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12:413-20.
    • (1995) Pharm Res , vol.12 , pp. 413-420
    • Amidon, G.L.1    Lennernäs, H.2    Shah, V.P.3    Crison, J.R.4
  • 172
    • 72749126885 scopus 로고    scopus 로고
    • Guidance for industry: Waiver of in vivo bioavailability and bioequivalence studies for imidiate-release solid oral dosage forms based on a biopharmaceutics classification system
    • CDER/FDA.
    • CDER/FDA. Guidance for industry: Waiver of in vivo bioavailability and bioequivalence studies for imidiate-release solid oral dosage forms based on a biopharmaceutics classification system. Center for Drug Evaluation and Research 2000.
    • (2000) Center for Drug Evaluation and Research
  • 173
    • 4644231090 scopus 로고    scopus 로고
    • Biopharmaceutic classification system: A scientific framework for pharmacokinetic optimization in drug research
    • 1:CAS:528:DC%2BD2cXnvFars7k%3D 15544432
    • Varma MV, Khandavilli S, Ashokraj Y, Jain A, Dhanikula A, Sood A, et al. Biopharmaceutic classification system: a scientific framework for pharmacokinetic optimization in drug research. Curr Drug Metab. 2004;5:375-88.
    • (2004) Curr Drug Metab , vol.5 , pp. 375-388
    • Varma, M.V.1    Khandavilli, S.2    Ashokraj, Y.3    Jain, A.4    Dhanikula, A.5    Sood, A.6
  • 174
    • 0031925268 scopus 로고    scopus 로고
    • Human intestinal permeability
    • 1:STN:280:DyaK1c3itV2gsw%3D%3D 9548891
    • Lennernas H. Human intestinal permeability. J Pharm Sci. 1998;87:403-10.
    • (1998) J Pharm Sci , vol.87 , pp. 403-410
    • Lennernas, H.1
  • 175
    • 0031780252 scopus 로고    scopus 로고
    • Jejunal absorption and metabolism of R/S-verapamil in humans
    • 1:CAS:528:DyaK1cXjvFWlsbk%3D 9647350
    • Sandstrom R, Karlsson A, Knutson L, Lennernas H. Jejunal absorption and metabolism of R/S-verapamil in humans. Pharm Res. 1998;15:856-62.
    • (1998) Pharm Res , vol.15 , pp. 856-862
    • Sandstrom, R.1    Karlsson, A.2    Knutson, L.3    Lennernas, H.4
  • 176
    • 35648978876 scopus 로고    scopus 로고
    • The use of BDDCS in classifying the permeability of marketed drugs
    • 3580995 1:CAS:528:DC%2BD1cXivFOitbs%3D 18236138
    • Benet LZ, Amidon GL, Barends DM, Lennernas H, Polli JE, Shah VP, et al. The use of BDDCS in classifying the permeability of marketed drugs. Pharm Res. 2008;25:483-8.
    • (2008) Pharm Res , vol.25 , pp. 483-488
    • Benet, L.Z.1    Amidon, G.L.2    Barends, D.M.3    Lennernas, H.4    Polli, J.E.5    Shah, V.P.6
  • 178
    • 84891583728 scopus 로고    scopus 로고
    • Absorption and drug development: Solubility, permeability, and charge state
    • Avdeef A. Absorption and drug development: solubility, permeability, and charge state. John Wiley & Sons, 2012.
    • (2012) John Wiley & Sons
    • Avdeef, A.1
  • 180
    • 84919799153 scopus 로고    scopus 로고
    • Practical permeability-based hepatic clearance classification system (HepCCS) in drug discovery
    • 1:CAS:528:DC%2BC2cXitFantLjK 25531965
    • Fan PW, Song Y, Berezhkovskiy LM, Cheong J, Plise EG, Khojasteh SC. Practical permeability-based hepatic clearance classification system (HepCCS) in drug discovery. Future Med Chem. 2014;6:1995-2012.
    • (2014) Future Med Chem , vol.6 , pp. 1995-2012
    • Fan, P.W.1    Song, Y.2    Berezhkovskiy, L.M.3    Cheong, J.4    Plise, E.G.5    Khojasteh, S.C.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.