메뉴 건너뛰기




Volumn 36, Issue 2, 2015, Pages 71-92

Dealing with the complex drug-drug interactions: Towards mechanistic models

Author keywords

CYP; drug drug interactions; metabolites; physiologically based pharmacokinetic model; transporters

Indexed keywords

CYTOCHROME P450; CYTOCHROME P450 2C19; CYTOCHROME P450 2C8; CYTOCHROME P450 2D6; CYTOCHROME P450 3A; CYTOCHROME P450 3A4; DRUG METABOLIZING ENZYME; CARRIER PROTEIN;

EID: 84924580597     PISSN: 01422782     EISSN: 1099081X     Source Type: Journal    
DOI: 10.1002/bdd.1934     Document Type: Review
Times cited : (60)

References (135)
  • 2
    • 0031794361 scopus 로고    scopus 로고
    • Inhibition and induction of cytochrome P450 and the clinical implications
    • Lin JH, Lu AY,. Inhibition and induction of cytochrome P450 and the clinical implications. Clin Pharmacokinet 1998; 35: 361-390.
    • (1998) Clin Pharmacokinet , vol.35 , pp. 361-390
    • Lin, J.H.1    Lu, A.Y.2
  • 3
    • 0037068964 scopus 로고    scopus 로고
    • Clinical importance of the cytochromes P450
    • Nebert DW, Russell DW,. Clinical importance of the cytochromes P450. Lancet 2002; 360: 1155-1162.
    • (2002) Lancet , vol.360 , pp. 1155-1162
    • Nebert, D.W.1    Russell, D.W.2
  • 4
    • 84861349018 scopus 로고    scopus 로고
    • Transporter-mediated drug - Drug interactions involving OATP substrates: Predictions based on in vitro inhibition studies
    • Yoshida K, Maeda K, Sugiyama Y,. Transporter-mediated drug-drug interactions involving OATP substrates: predictions based on in vitro inhibition studies. Clin Pharmacol Ther 2012; 91: 1053-1064.
    • (2012) Clin Pharmacol Ther , vol.91 , pp. 1053-1064
    • Yoshida, K.1    Maeda, K.2    Sugiyama, Y.3
  • 5
    • 0032924934 scopus 로고    scopus 로고
    • Cytochrome P-450 3A4: Regulation and role in drug metabolism
    • Guengerich FP,. Cytochrome P-450 3A4: regulation and role in drug metabolism. Annu Rev Pharmacol Toxicol 1999; 39: 1-17.
    • (1999) Annu Rev Pharmacol Toxicol , vol.39 , pp. 1-17
    • Guengerich, F.P.1
  • 6
    • 0031723235 scopus 로고    scopus 로고
    • Prediction of pharmacokinetic alterations caused by drug-drug interactions: Metabolic interaction in the liver
    • Ito K, Iwatsubo T, Kanamitsu S, Ueda K, Suzuki H, Sugiyama Y,. Prediction of pharmacokinetic alterations caused by drug-drug interactions: metabolic interaction in the liver. Pharmacol Rev 1998; 50: 387-412.
    • (1998) Pharmacol Rev , vol.50 , pp. 387-412
    • Ito, K.1    Iwatsubo, T.2    Kanamitsu, S.3    Ueda, K.4    Suzuki, H.5    Sugiyama, Y.6
  • 8
    • 18844369894 scopus 로고    scopus 로고
    • Impact of parallel pathways of drug elimination and multiple cytochrome P450 involvement on drug-drug interactions: CYP2D6 paradigm
    • Ito K, Hallifax D, Obach RS, Houston JB,. Impact of parallel pathways of drug elimination and multiple cytochrome P450 involvement on drug-drug interactions: CYP2D6 paradigm. Drug Metab Dispos 2005; 33: 837-844.
    • (2005) Drug Metab Dispos , vol.33 , pp. 837-844
    • Ito, K.1    Hallifax, D.2    Obach, R.S.3    Houston, J.B.4
  • 9
    • 27644596457 scopus 로고    scopus 로고
    • Predicting in vivo drug interactions from in vitro drug discovery data
    • Wienkers LC, Heath TG,. Predicting in vivo drug interactions from in vitro drug discovery data. Nature Rev Drug Discov 2005; 4: 825-833.
    • (2005) Nature Rev Drug Discov , vol.4 , pp. 825-833
    • Wienkers, L.C.1    Heath, T.G.2
  • 10
    • 84888632710 scopus 로고    scopus 로고
    • A perspective on the prediction of drug pharmacokinetics and disposition in drug research and development
    • Di L, Feng B, Goosen TC, et al., A perspective on the prediction of drug pharmacokinetics and disposition in drug research and development. Drug Metab Dispos 2013; 41: 1975-1993.
    • (2013) Drug Metab Dispos , vol.41 , pp. 1975-1993
    • Di, L.1    Feng, B.2    Goosen, T.C.3
  • 11
    • 67650825002 scopus 로고    scopus 로고
    • Comparison of different algorithms for predicting clinical drug-drug interactions, based on the use of CYP3A4 in vitro data: Predictions of compounds as precipitants of interaction
    • Fahmi OA, Hurst S, Plowchalk D, et al., Comparison of different algorithms for predicting clinical drug-drug interactions, based on the use of CYP3A4 in vitro data: predictions of compounds as precipitants of interaction. Drug Metab Dispos 2009; 37: 1658-1666.
    • (2009) Drug Metab Dispos , vol.37 , pp. 1658-1666
    • Fahmi, O.A.1    Hurst, S.2    Plowchalk, D.3
  • 12
    • 47949115175 scopus 로고    scopus 로고
    • A combined model for predicting CYP3A4 clinical net drug-drug interaction based on CYP3A4 inhibition, inactivation, and induction determined in vitro
    • Fahmi OA, Maurer TS, Kish M, Cardenas E, Boldt S, Nettleton D,. A combined model for predicting CYP3A4 clinical net drug-drug interaction based on CYP3A4 inhibition, inactivation, and induction determined in vitro. Drug Metab Dispos 2008; 36: 1698-1708.
    • (2008) Drug Metab Dispos , vol.36 , pp. 1698-1708
    • Fahmi, O.A.1    Maurer, T.S.2    Kish, M.3    Cardenas, E.4    Boldt, S.5    Nettleton, D.6
  • 13
    • 33947587274 scopus 로고    scopus 로고
    • Simulation and prediction of in vivo drug metabolism in human populations from in vitro data
    • Rostami-Hodjegan A, Tucker GT,. Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 2007; 6: 140-148.
    • (2007) Nat Rev Drug Discov , vol.6 , pp. 140-148
    • Rostami-Hodjegan, A.1    Tucker, G.T.2
  • 14
    • 84858660647 scopus 로고    scopus 로고
    • From preclinical to human - Prediction of oral absorption and drug-drug interaction potential using physiologically based pharmacokinetic (PBPK) modeling approach in an industrial setting: A workflow by using case example
    • Sinha VK, Snoeys J, Osselaer NV, Peer AV, Mackie C, Heald D,. From preclinical to human-prediction of oral absorption and drug-drug interaction potential using physiologically based pharmacokinetic (PBPK) modeling approach in an industrial setting: a workflow by using case example. Biopharm Drug Dispos 2012; 33: 111-121.
    • (2012) Biopharm Drug Dispos , vol.33 , pp. 111-121
    • Sinha, V.K.1    Snoeys, J.2    Osselaer, N.V.3    Peer, A.V.4    Mackie, C.5    Heald, D.6
  • 15
    • 84893740148 scopus 로고    scopus 로고
    • Evaluation of various static in vitro-in vivo extrapolation models for risk assessment of the CYP3A inhibition potential of an investigational drug
    • Vieira ML, Kirby B, Ragueneau-Majlessi I, et al., Evaluation of various static in vitro-in vivo extrapolation models for risk assessment of the CYP3A inhibition potential of an investigational drug. Clin Pharmacol Ther 2014; 95: 189-198.
    • (2014) Clin Pharmacol Ther , vol.95 , pp. 189-198
    • Vieira, M.L.1    Kirby, B.2    Ragueneau-Majlessi, I.3
  • 16
    • 84875160515 scopus 로고    scopus 로고
    • Model-based approaches to predict drug-drug interactions associated with hepatic uptake transporters: Preclinical, clinical and beyond
    • Barton HA, Lai Y, Goosen TC, 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: 459-472.
    • (2013) Expert Opin Drug Metab Toxicol , vol.9 , pp. 459-472
    • Barton, H.A.1    Lai, Y.2    Goosen, T.C.3
  • 17
    • 79951765993 scopus 로고    scopus 로고
    • Physiologically-based pharmacokinetics in drug development and regulatory science
    • Rowland M, Peck C, Tucker G,. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol 2011; 51: 45-73.
    • (2011) Annu Rev Pharmacol Toxicol , vol.51 , pp. 45-73
    • Rowland, M.1    Peck, C.2    Tucker, G.3
  • 18
    • 84921297240 scopus 로고    scopus 로고
    • USFDA Implications for Dosing, and Labeling Recommendations. Center for Drug Evaluation and Research (CDER)
    • USFDA. Drug Interaction Studies-Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. Center for Drug Evaluation and Research (CDER) 2012.
    • (2012) Drug Interaction Studies - Study Design, Data Analysis
  • 19
    • 84899442309 scopus 로고    scopus 로고
    • PBPK model describes the effects of comedication and genetic polymorphism on systemic exposure of drugs that undergo multiple clearance pathways
    • Vieira M, Kim M, Apparaju S, et al., PBPK model describes the effects of comedication and genetic polymorphism on systemic exposure of drugs that undergo multiple clearance pathways. Clin Pharmacol Ther 2014; 95: 550-557.
    • (2014) Clin Pharmacol Ther , vol.95 , pp. 550-557
    • Vieira, M.1    Kim, M.2    Apparaju, S.3
  • 20
    • 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
    • Fenner KS, Jones HM, Ullah M, 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
  • 21
    • 70350048926 scopus 로고    scopus 로고
    • Impact of OATP transporters on pharmacokinetics
    • 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
  • 22
    • 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
    • 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
  • 23
    • 33644847684 scopus 로고    scopus 로고
    • Transporters as a determinant of drug clearance and tissue distribution
    • Shitara Y, Horie T, Sugiyama Y,. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci 2006; 27: 425-446.
    • (2006) Eur J Pharm Sci , vol.27 , pp. 425-446
    • Shitara, Y.1    Horie, T.2    Sugiyama, Y.3
  • 24
    • 80052968100 scopus 로고    scopus 로고
    • Identification of the rate-determining process in the hepatic clearance of atorvastatin in a clinical cassette microdosing study
    • Maeda K, Ikeda Y, Fujita T, 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: 575-581.
    • (2011) Clin Pharmacol Ther , vol.90 , pp. 575-581
    • Maeda, K.1    Ikeda, Y.2    Fujita, T.3
  • 25
    • 84906508843 scopus 로고    scopus 로고
    • Pitavastatin is a more sensitive and selective OATP1B clinical probe than rosuvastatin
    • Prueksaritanont T, Chu X, Evers R, et al., Pitavastatin is a more sensitive and selective OATP1B clinical probe than rosuvastatin. Br J Clin Pharmacol 2014; 78: 587-598.
    • (2014) Br J Clin Pharmacol , vol.78 , pp. 587-598
    • Prueksaritanont, T.1    Chu, X.2    Evers, R.3
  • 26
    • 49949104757 scopus 로고    scopus 로고
    • SLCO1B1 variants and statin-induced myopathy - A genomewide study
    • Group SC, Link E, Parish S, et al., SLCO1B1 variants and statin-induced myopathy-a genomewide study. N Engl J Med 2008; 359: 789-799.
    • (2008) N Engl J Med , vol.359 , pp. 789-799
    • Group, S.C.1    Link, E.2    Parish, S.3
  • 27
    • 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
    • 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. Expert Opin Drug Metab Toxicol 2009; 5: 703-729.
    • (2009) Expert Opin Drug Metab Toxicol , vol.5 , pp. 703-729
    • Ieiri, I.1    Higuchi, S.2    Sugiyama, Y.3
  • 28
    • 18744388590 scopus 로고    scopus 로고
    • Acute effects of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype 17
    • Niemi M, Neuvonen PJ, Hofmann U, et al., Acute effects of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype 17. Pharmacogenet Genomics 2005; 15: 303-309.
    • (2005) Pharmacogenet Genomics , vol.15 , pp. 303-309
    • Niemi, M.1    Neuvonen, P.J.2    Hofmann, U.3
  • 29
    • 0038209381 scopus 로고    scopus 로고
    • Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: Consequences for pravastatin pharmacokinetics
    • Nishizato Y, Ieiri I, Suzuki H, et al., Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: consequences for pravastatin pharmacokinetics. Clin Pharmacol Ther 2003; 73: 554-565.
    • (2003) Clin Pharmacol Ther , vol.73 , pp. 554-565
    • Nishizato, Y.1    Ieiri, I.2    Suzuki, H.3
  • 30
    • 84872229531 scopus 로고    scopus 로고
    • Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: Their roles in hepatic clearance and intestinal absorption
    • 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
  • 31
    • 71949127231 scopus 로고    scopus 로고
    • Interplay of transporters and enzymes in drug and metabolite processing
    • Pang KS, Maeng HJ, Fan J,. Interplay of transporters and enzymes in drug and metabolite processing. Mol Pharm 2009; 6: 1734-1755.
    • (2009) Mol Pharm , vol.6 , pp. 1734-1755
    • Pang, K.S.1    Maeng, H.J.2    Fan, J.3
  • 32
    • 84872234822 scopus 로고    scopus 로고
    • Hepatic and intestinal drug transporters: Prediction of pharmacokinetic effects caused by drug-drug interactions and genetic polymorphisms
    • 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
  • 33
    • 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 Dispos 2005; 33: 1-9.
    • (2005) Drug Metab Dispos , vol.33 , pp. 1-9
    • Liu, L.1    Pang, K.S.2
  • 34
    • 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
    • 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: 625-653.
    • (1977) J Pharmacokinet Biopharm , vol.5 , pp. 625-653
    • Pang, K.S.1    Rowland, M.2
  • 35
    • 0021798973 scopus 로고
    • Hepatic elimination - Dispersion model
    • Roberts M, Rowland M,. Hepatic elimination-dispersion model. J Pharm Sci 1985; 74: 585-587.
    • (1985) J Pharm Sci , vol.74 , pp. 585-587
    • Roberts, M.1    Rowland, M.2
  • 36
    • 0022552317 scopus 로고
    • A dispersion model of hepatic elimination: 2. Steady-state considerations - Influence of hepatic blood flow, binding within blood, and hepatocellular enzyme activity
    • Roberts MS, Rowland M,. A dispersion model of hepatic elimination: 2. Steady-state considerations-influence of hepatic blood flow, binding within blood, and hepatocellular enzyme activity. J Pharmacokinet Biopharm 1986; 14: 261-288.
    • (1986) J Pharmacokinet Biopharm , vol.14 , pp. 261-288
    • Roberts, M.S.1    Rowland, M.2
  • 37
    • 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
    • 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: 179-194.
    • (2012) Biopharm Drug Dispos , vol.33 , pp. 179-194
    • Camenisch, G.1    Umehara, K.2
  • 38
    • 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
    • Varma MV, Lin J, Bi YA, 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 Dispos 2013; 41: 966-974.
    • (2013) Drug Metab Dispos , vol.41 , pp. 966-974
    • Varma, M.V.1    Lin, J.2    Bi, Y.A.3
  • 39
    • 84879416924 scopus 로고    scopus 로고
    • And International Transporter C. ITC recommendations for transporter kinetic parameter estimation and translational modeling of transport-mediated PK and DDIs in humans
    • Zamek-Gliszczynski MJ, Lee CA, Poirier A, et al., and International Transporter C. ITC recommendations for transporter kinetic parameter estimation and translational modeling of transport-mediated PK and DDIs in humans. Clin Pharmacol Ther 2013; 94: 64-79.
    • (2013) Clin Pharmacol Ther , vol.94 , pp. 64-79
    • Zamek-Gliszczynski, M.J.1    Lee, C.A.2    Poirier, A.3
  • 40
    • 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 Dispos 2007; 35: 1766-1780.
    • (2007) Drug Metab Dispos , 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
  • 41
    • 79953296456 scopus 로고    scopus 로고
    • Simulation of human intravenous and oral pharmacokinetics of 21 diverse compounds using physiologically based pharmacokinetic modelling
    • Jones HM, Gardner IB, Collard WT, et al., Simulation of human intravenous and oral pharmacokinetics of 21 diverse compounds using physiologically based pharmacokinetic modelling. Clin Pharmacokinet 2011; 50: 331-347.
    • (2011) Clin Pharmacokinet , vol.50 , pp. 331-347
    • Jones, H.M.1    Gardner, I.B.2    Collard, W.T.3
  • 43
    • 84878723655 scopus 로고    scopus 로고
    • Disposition pathway-dependent approach for predicting organic anion-transporting polypeptide-mediated drug-drug interactions
    • Hu ZY,. Disposition pathway-dependent approach for predicting organic anion-transporting polypeptide-mediated drug-drug interactions. Clin Pharmacokinet 2013; 52: 433-441.
    • (2013) Clin Pharmacokinet , vol.52 , pp. 433-441
    • Hu, Z.Y.1
  • 44
    • 79951768576 scopus 로고    scopus 로고
    • Application of physiologically based pharmacokinetic modeling and clearance concept to drugs showing transporter-mediated distribution and clearance in humans
    • Watanabe T, Kusuhara H, Sugiyama Y,. Application of physiologically based pharmacokinetic modeling and clearance concept to drugs showing transporter-mediated distribution and clearance in humans. J Pharmacokinet Pharmacodyn 2010; 37: 575-590.
    • (2010) J Pharmacokinet Pharmacodyn , vol.37 , pp. 575-590
    • Watanabe, T.1    Kusuhara, H.2    Sugiyama, Y.3
  • 46
    • 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; 29: 2860-2873.
    • (2012) Pharm Res , vol.29 , pp. 2860-2873
    • Varma, M.V.1    Lai, Y.2    Feng, B.3    Litchfield, J.4    Goosen, T.C.5    Bergman, A.6
  • 47
    • 84876483882 scopus 로고    scopus 로고
    • Mechanistic modeling to predict the transporter- and enzyme-mediated drug-drug interactions of repaglinide
    • 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: 1188-1199.
    • (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
  • 48
    • 0033994866 scopus 로고    scopus 로고
    • Quantitative prediction of in vivo drug-drug interactions from in vitro data based on physiological pharmacokinetics: Use of maximum unbound concentration of inhibitor at the inlet to the liver
    • Kanamitsu S, Ito K, Sugiyama Y,. Quantitative prediction of in vivo drug-drug interactions from in vitro data based on physiological pharmacokinetics: use of maximum unbound concentration of inhibitor at the inlet to the liver. Pharm Res 2000; 17: 336-343.
    • (2000) Pharm Res , vol.17 , pp. 336-343
    • Kanamitsu, S.1    Ito, K.2    Sugiyama, Y.3
  • 49
    • 84859346272 scopus 로고    scopus 로고
    • In vitro and in silico strategies to identify OATP1B1 inhibitors and predict clinical drug-drug interactions
    • Karlgren M, Ahlin G, Bergstrom CA, Svensson R, Palm J, Artursson P,. In vitro and in silico strategies to identify OATP1B1 inhibitors and predict clinical drug-drug interactions. Pharm Res 2012; 29: 411-426.
    • (2012) Pharm Res , vol.29 , pp. 411-426
    • Karlgren, M.1    Ahlin, G.2    Bergstrom, C.A.3    Svensson, R.4    Palm, J.5    Artursson, P.6
  • 50
    • 84861494749 scopus 로고    scopus 로고
    • Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): Influence of protein expression on drug-drug interactions
    • Karlgren M, Vildhede A, Norinder U, et al., Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions. J Med Chem 2012; 55: 4740-4763.
    • (2012) J Med Chem , vol.55 , pp. 4740-4763
    • Karlgren, M.1    Vildhede, A.2    Norinder, U.3
  • 51
    • 80054680414 scopus 로고    scopus 로고
    • Pharmacokinetics of voriconazole administered concomitantly with fluconazole and population-based simulation for sequential use
    • Damle B, Varma MV, Wood N,. Pharmacokinetics of voriconazole administered concomitantly with fluconazole and population-based simulation for sequential use. Antimicrob Agents Chemother 2011; 55: 5172-5177.
    • (2011) Antimicrob Agents Chemother , vol.55 , pp. 5172-5177
    • Damle, B.1    Varma, M.V.2    Wood, N.3
  • 52
    • 78650016675 scopus 로고    scopus 로고
    • Assessment of algorithms for predicting drug-drug interactions via inhibition mechanisms: Comparison of dynamic and static models
    • Guest EJ, Rowland-Yeo K, Rostami-Hodjegan A, Tucker GT, Houston JB, Galetin A,. Assessment of algorithms for predicting drug-drug interactions via inhibition mechanisms: comparison of dynamic and static models. Br J Clin Pharmacol 2011; 71: 72-87.
    • (2011) Br J Clin Pharmacol , vol.71 , pp. 72-87
    • Guest, E.J.1    Rowland-Yeo, K.2    Rostami-Hodjegan, A.3    Tucker, G.T.4    Houston, J.B.5    Galetin, A.6
  • 53
    • 84863947716 scopus 로고    scopus 로고
    • Evaluation of the use of static and dynamic models to predict drug-drug interaction and its associated variability: Impact on drug discovery and early development
    • Peters SA, Schroeder PE, Giri N, Dolgos H,. Evaluation of the use of static and dynamic models to predict drug-drug interaction and its associated variability: impact on drug discovery and early development. Drug Metab Dispos 2012; 40: 1495-1507.
    • (2012) Drug Metab Dispos , vol.40 , pp. 1495-1507
    • Peters, S.A.1    Schroeder, P.E.2    Giri, N.3    Dolgos, H.4
  • 54
    • 84903633472 scopus 로고    scopus 로고
    • Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide
    • Varma MV, Scialis R, Lin J, et al., Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide. AAPS Journal 2014; 16: 736-748.
    • (2014) AAPS Journal , vol.16 , pp. 736-748
    • Varma, M.V.1    Scialis, R.2    Lin, J.3
  • 55
    • 84859910816 scopus 로고    scopus 로고
    • Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data
    • Jones HM, Barton HA, Lai Y, et al., Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data. Drug Metab Dispos 2012; 40: 1007-1017.
    • (2012) Drug Metab Dispos , vol.40 , pp. 1007-1017
    • Jones, H.M.1    Barton, H.A.2    Lai, Y.3
  • 56
    • 84904201004 scopus 로고    scopus 로고
    • A 'middle-out' approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling
    • Li R, Barton HA, Yates PD, et al., A 'middle-out' approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn 2014; 41: 197-209.
    • (2014) J Pharmacokinet Pharmacodyn , vol.41 , pp. 197-209
    • Li, R.1    Barton, H.A.2    Yates, P.D.3
  • 57
    • 84891867228 scopus 로고    scopus 로고
    • A mechanistic framework for in vitro-in vivo extrapolation of liver membrane transporters: Prediction of drug-drug interaction between rosuvastatin and cyclosporine
    • Jamei M, Bajot F, Neuhoff S, et al., A mechanistic framework for in vitro-in vivo extrapolation of liver membrane transporters: prediction of drug-drug interaction between rosuvastatin and cyclosporine. Clin Pharmacokinet 2014; 53: 73-87.
    • (2014) Clin Pharmacokinet , vol.53 , pp. 73-87
    • Jamei, M.1    Bajot, F.2    Neuhoff, S.3
  • 58
    • 84903633472 scopus 로고    scopus 로고
    • Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide
    • Varma MV, Scialis RJ, Lin J, et al., Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide. AAPS J 2014; 16: 736-748.
    • (2014) AAPS J , vol.16 , pp. 736-748
    • Varma, M.V.1    Scialis, R.J.2    Lin, J.3
  • 59
    • 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
    • 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: 214-223.
    • (2014) J Pharmacol Exp Ther , vol.351 , pp. 214-223
    • Varma, M.V.1    Bi, Y.A.2    Kimoto, E.3    Lin, J.4
  • 60
    • 0042318871 scopus 로고    scopus 로고
    • CYP2C8 and CYP3A4 are the principal enzymes involved in the human in vitro biotransformation of the insulin secretagogue repaglinide
    • Bidstrup TB, Bjornsdottir I, Sidelmann UG, Thomsen MS, Hansen KT,. CYP2C8 and CYP3A4 are the principal enzymes involved in the human in vitro biotransformation of the insulin secretagogue repaglinide. Br J Clin Pharmacol 2003; 56: 305-314.
    • (2003) Br J Clin Pharmacol , vol.56 , pp. 305-314
    • Bidstrup, T.B.1    Bjornsdottir, I.2    Sidelmann, U.G.3    Thomsen, M.S.4    Hansen, K.T.5
  • 61
    • 25844448609 scopus 로고    scopus 로고
    • Metabolism of repaglinide by CYP2C8 and CYP3A4 in vitro: Effect of fibrates and rifampicin
    • Kajosaari LI, Laitila J, Neuvonen PJ, Backman JT,. Metabolism of repaglinide by CYP2C8 and CYP3A4 in vitro: effect of fibrates and rifampicin. Basic Clin Pharmacol Toxicol 2005; 97: 249-256.
    • (2005) Basic Clin Pharmacol Toxicol , vol.97 , pp. 249-256
    • Kajosaari, L.I.1    Laitila, J.2    Neuvonen, P.J.3    Backman, J.T.4
  • 62
    • 84862658556 scopus 로고    scopus 로고
    • A comprehensive assessment of repaglinide metabolic pathways: Impact of choice of in vitro system and relative enzyme contribution to in vitro clearance
    • Sall C, Houston JB, Galetin A,. A comprehensive assessment of repaglinide metabolic pathways: impact of choice of in vitro system and relative enzyme contribution to in vitro clearance. Drug Metab Dispos 2012; 40: 1279-1289.
    • (2012) Drug Metab Dispos , vol.40 , pp. 1279-1289
    • Sall, C.1    Houston, J.B.2    Galetin, A.3
  • 64
    • 20444466590 scopus 로고    scopus 로고
    • Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics
    • Niemi M, Backman JT, Kajosaari LI, et al., Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics. Clin Pharmacol Ther 2005; 77: 468-478.
    • (2005) Clin Pharmacol Ther , vol.77 , pp. 468-478
    • Niemi, M.1    Backman, J.T.2    Kajosaari, L.I.3
  • 65
    • 29944446317 scopus 로고    scopus 로고
    • Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: Implications for drug-drug interactions
    • Ogilvie BW, Zhang D, Li W, et al., Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: implications for drug-drug interactions. Drug Metab Dispos 2006; 34: 191-197.
    • (2006) Drug Metab Dispos , vol.34 , pp. 191-197
    • Ogilvie, B.W.1    Zhang, D.2    Li, W.3
  • 66
    • 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
    • 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: 228-236.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 228-236
    • Shitara, Y.1    Hirano, M.2    Sato, H.3    Sugiyama, Y.4
  • 68
    • 42049097101 scopus 로고    scopus 로고
    • Multiple inhibition mechanisms and prediction of drug-drug interactions: Status of metabolism and transporter models as exemplified by gemfibrozil-drug interactions
    • Hinton LK, Galetin A, Houston JB,. Multiple inhibition mechanisms and prediction of drug-drug interactions: status of metabolism and transporter models as exemplified by gemfibrozil-drug interactions. Pharm Res 2008; 25: 1063-1074.
    • (2008) Pharm Res , vol.25 , pp. 1063-1074
    • Hinton, L.K.1    Galetin, A.2    Houston, J.B.3
  • 69
    • 80053146805 scopus 로고    scopus 로고
    • Dose-dependent interaction between gemfibrozil and repaglinide in humans: Strong inhibition of CYP2C8 with subtherapeutic gemfibrozil doses
    • Honkalammi J, Niemi M, Neuvonen PJ, Backman JT,. Dose-dependent interaction between gemfibrozil and repaglinide in humans: strong inhibition of CYP2C8 with subtherapeutic gemfibrozil doses. Drug Metab Dispos 2011; 39: 1977-1986.
    • (2011) Drug Metab Dispos , vol.39 , pp. 1977-1986
    • Honkalammi, J.1    Niemi, M.2    Neuvonen, P.J.3    Backman, J.T.4
  • 70
    • 84860014528 scopus 로고    scopus 로고
    • Gemfibrozil is a strong inactivator of CYP2C8 in very small multiple doses
    • Honkalammi J, Niemi M, Neuvonen PJ, Backman JT,. Gemfibrozil is a strong inactivator of CYP2C8 in very small multiple doses. Clin Pharmacol Ther 2012; 91: 846-855.
    • (2012) Clin Pharmacol Ther , vol.91 , pp. 846-855
    • Honkalammi, J.1    Niemi, M.2    Neuvonen, P.J.3    Backman, J.T.4
  • 71
    • 0037627707 scopus 로고    scopus 로고
    • Effects of gemfibrozil, itraconazole, and their combination on the pharmacokinetics and pharmacodynamics of repaglinide: Potentially hazardous interaction between gemfibrozil and repaglinide
    • Niemi M, Backman JT, Neuvonen M, Neuvonen PJ,. Effects of gemfibrozil, itraconazole, and their combination on the pharmacokinetics and pharmacodynamics of repaglinide: potentially hazardous interaction between gemfibrozil and repaglinide. Diabetologia 2003; 46: 347-351.
    • (2003) Diabetologia , vol.46 , pp. 347-351
    • Niemi, M.1    Backman, J.T.2    Neuvonen, M.3    Neuvonen, P.J.4
  • 72
    • 84872705391 scopus 로고    scopus 로고
    • Analysis of the repaglinide concentration increase produced by gemfibrozil and itraconazole based on the inhibition of the hepatic uptake transporter and metabolic enzymes
    • Kudo T, Hisaka A, Sugiyama Y, Ito K,. Analysis of the repaglinide concentration increase produced by gemfibrozil and itraconazole based on the inhibition of the hepatic uptake transporter and metabolic enzymes. Drug Metab Dispos 2013; 41: 362-371.
    • (2013) Drug Metab Dispos , vol.41 , pp. 362-371
    • Kudo, T.1    Hisaka, A.2    Sugiyama, Y.3    Ito, K.4
  • 73
    • 84895776723 scopus 로고    scopus 로고
    • Reduced physiologically-based pharmacokinetic model of repaglinide: Impact of OATP1B1 and CYP2C8 genotype and source of in vitro data on the prediction of drug-drug interaction risk
    • Gertz M, Tsamandouras N, Sall C, Houston JB, Galetin A,. Reduced physiologically-based pharmacokinetic model of repaglinide: impact of OATP1B1 and CYP2C8 genotype and source of in vitro data on the prediction of drug-drug interaction risk. Pharm Res 2014; 31: 2367-2382.
    • (2014) Pharm Res , vol.31 , pp. 2367-2382
    • Gertz, M.1    Tsamandouras, N.2    Sall, C.3    Houston, J.B.4    Galetin, A.5
  • 74
    • 0345275778 scopus 로고    scopus 로고
    • Modeling of intestinal drug absorption: Roles of transporters and metabolic enzymes (for the Gillette Review Series)
    • Pang KS,. Modeling of intestinal drug absorption: roles of transporters and metabolic enzymes (for the Gillette Review Series). Drug Metab Dispos 2003; 31: 1507-1519.
    • (2003) Drug Metab Dispos , vol.31 , pp. 1507-1519
    • Pang, K.S.1
  • 75
    • 0345275777 scopus 로고    scopus 로고
    • The small intestine as a xenobiotic-metabolizing organ
    • Kaminsky LS, Zhang Q-Y,. The small intestine as a xenobiotic-metabolizing organ. Drug Metab Dispos 2003; 31: 1520-1525.
    • (2003) Drug Metab Dispos , vol.31 , pp. 1520-1525
    • Kaminsky, L.S.1    Zhang, Q.-Y.2
  • 76
    • 77957130129 scopus 로고    scopus 로고
    • Extensive intestinal first-pass elimination and predominant hepatic distribution of berberine explain its low plasma levels in rats
    • Liu Y-T, Hao H-P, Xie H-G, et al., Extensive intestinal first-pass elimination and predominant hepatic distribution of berberine explain its low plasma levels in rats. Drug Metab Dispos 2010; 38: 1779-1784.
    • (2010) Drug Metab Dispos , vol.38 , pp. 1779-1784
    • Liu, Y.-T.1    Hao, H.-P.2    Xie, H.-G.3
  • 78
    • 1542376174 scopus 로고    scopus 로고
    • Cytochrome P450 3A4 and P-glycoprotein expression in human small intestinal enterocytes and hepatocytes: A comparative analysis in paired tissue specimens
    • Von Richter O, Burk O, Fromm MF, Thon KP, Eichelbaum M, Kivistö KT,. Cytochrome P450 3A4 and P-glycoprotein expression in human small intestinal enterocytes and hepatocytes: a comparative analysis in paired tissue specimens. Clin Pharmacol Ther 2004; 75: 172-183.
    • (2004) Clin Pharmacol Ther , vol.75 , pp. 172-183
    • Von Richter, O.1    Burk, O.2    Fromm, M.F.3    Thon, K.P.4    Eichelbaum, M.5    Kivistö, K.T.6
  • 79
    • 0031445547 scopus 로고    scopus 로고
    • Characterization of interintestinal and intraintestinal variations in human CYP3A-dependent metabolism
    • Paine MF, Khalighi M, Fisher JM, et al., Characterization of interintestinal and intraintestinal variations in human CYP3A-dependent metabolism. J Pharmacol Exp Ther 1997; 283: 1552-1562.
    • (1997) J Pharmacol Exp Ther , vol.283 , pp. 1552-1562
    • Paine, M.F.1    Khalighi, M.2    Fisher, J.M.3
  • 80
    • 33747599176 scopus 로고    scopus 로고
    • Intestinal and hepatic metabolic activity of five cytochrome P450 enzymes: Impact on prediction of first-pass metabolism
    • Galetin A, Houston JB,. Intestinal and hepatic metabolic activity of five cytochrome P450 enzymes: impact on prediction of first-pass metabolism. J Pharmacol Exp Ther 2006; 318: 1220-1229.
    • (2006) J Pharmacol Exp Ther , vol.318 , pp. 1220-1229
    • Galetin, A.1    Houston, J.B.2
  • 81
    • 34247352929 scopus 로고    scopus 로고
    • Gene and protein expression of P-glycoprotein, MRP1, MRP2, and CYP3A4 in the small and large human intestine
    • Berggren S, Gall C, Wollnitz N, et al., Gene and protein expression of P-glycoprotein, MRP1, MRP2, and CYP3A4 in the small and large human intestine. Mol Pharm 2007; 4: 252-257.
    • (2007) Mol Pharm , vol.4 , pp. 252-257
    • Berggren, S.1    Gall, C.2    Wollnitz, N.3
  • 82
    • 33750181267 scopus 로고    scopus 로고
    • Regional levels of drug transporters along the human intestinal tract: Co-expression of ABC and SLC transporters and comparison with Caco-2 cells
    • Englund G, Rorsman F, Rönnblom A, et al., Regional levels of drug transporters along the human intestinal tract: co-expression of ABC and SLC transporters and comparison with Caco-2 cells. Eur J Pharm Sci 2006; 29: 269-277.
    • (2006) Eur J Pharm Sci , vol.29 , pp. 269-277
    • Englund, G.1    Rorsman, F.2    Rönnblom, A.3
  • 83
    • 0036225166 scopus 로고    scopus 로고
    • Intestinal metabolism promotes regional differences in apical uptake of indinavir: Coupled effect of P-glycoprotein and cytochrome P450 3A on indinavir membrane permeability in rat
    • Li LY, Amidon GL, Kim JS, et al., Intestinal metabolism promotes regional differences in apical uptake of indinavir: coupled effect of P-glycoprotein and cytochrome P450 3A on indinavir membrane permeability in rat. J Pharmacol Exp Ther 2002; 301: 586-593.
    • (2002) J Pharmacol Exp Ther , vol.301 , pp. 586-593
    • Li, L.Y.1    Amidon, G.L.2    Kim, J.S.3
  • 84
    • 33747821713 scopus 로고    scopus 로고
    • P-glycoprotein and an unstirred water layer barring digoxin absorption in the vascularly perfused rat small intestine preparation: Induction studies with pregnenolone-16α-carbonitrile
    • Liu S, Tam D, Chen X, Pang KS,. P-glycoprotein and an unstirred water layer barring digoxin absorption in the vascularly perfused rat small intestine preparation: induction studies with pregnenolone-16α-carbonitrile. Drug Metab Dispos 2006; 34: 1468-1479.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1468-1479
    • Liu, S.1    Tam, D.2    Chen, X.3    Pang, K.S.4
  • 85
    • 0142010611 scopus 로고    scopus 로고
    • P-glycoprotein increases from proximal to distal regions of human small intestine
    • Mouly S, Paine MF,. P-glycoprotein increases from proximal to distal regions of human small intestine. Pharm Res 2003; 20: 1595-1599.
    • (2003) Pharm Res , vol.20 , pp. 1595-1599
    • Mouly, S.1    Paine, M.F.2
  • 86
    • 0141569628 scopus 로고    scopus 로고
    • Expression profiles of drug-metabolizing enzyme CYP3A and drug efflux transporter multidrug resistance 1 subfamily mRNAs in rat small intestine
    • Takara K, Ohnishi N, Horibe S, Yokoyama T,. Expression profiles of drug-metabolizing enzyme CYP3A and drug efflux transporter multidrug resistance 1 subfamily mRNAs in rat small intestine. Drug Metab Dispos 2003; 31: 1235-1239.
    • (2003) Drug Metab Dispos , vol.31 , pp. 1235-1239
    • Takara, K.1    Ohnishi, N.2    Horibe, S.3    Yokoyama, T.4
  • 87
    • 77957735036 scopus 로고    scopus 로고
    • Declèves X, Bouzom F, et al. Effect of variations in the amounts of P-glycoprotein (ABCB1), BCRP (ABCG2) and CYP3A4 along the human small intestine on PBPK models for predicting intestinal first pass
    • Bruyere A,. Declèves X, Bouzom F, et al. Effect of variations in the amounts of P-glycoprotein (ABCB1), BCRP (ABCG2) and CYP3A4 along the human small intestine on PBPK models for predicting intestinal first pass. Mol Pharm 2010; 7: 1596-1607.
    • (2010) Mol Pharm , vol.7 , pp. 1596-1607
    • Bruyere, A.1
  • 88
    • 67049114269 scopus 로고    scopus 로고
    • Methodology for development of a physiological model incorporating CYP3A and P-glycoprotein for the prediction of intestinal drug absorption
    • Badhan R, Penny J, Galetin A, Houston JB,. Methodology for development of a physiological model incorporating CYP3A and P-glycoprotein for the prediction of intestinal drug absorption. J Pharm Sci 2009; 98: 2180-2197.
    • (2009) J Pharm Sci , vol.98 , pp. 2180-2197
    • Badhan, R.1    Penny, J.2    Galetin, A.3    Houston, J.B.4
  • 89
    • 77953737073 scopus 로고    scopus 로고
    • Prediction of human intestinal first-pass metabolism of 25 CYP3A substrates from in vitro clearance and permeability data
    • Gertz M, Harrison A, Houston JB, Galetin A,. Prediction of human intestinal first-pass metabolism of 25 CYP3A substrates from in vitro clearance and permeability data. Drug Metab Dispos 2010; 38: 1147-1158.
    • (2010) Drug Metab Dispos , vol.38 , pp. 1147-1158
    • Gertz, M.1    Harrison, A.2    Houston, J.B.3    Galetin, A.4
  • 90
    • 84871294584 scopus 로고    scopus 로고
    • Why we need proper PBPK models to examine intestine and liver oral drug absorption
    • Chow E, Sandy PK,. Why we need proper PBPK models to examine intestine and liver oral drug absorption. Curr Drug Metab 2013; 14: 57-79.
    • (2013) Curr Drug Metab , vol.14 , pp. 57-79
    • Chow, E.1    Sandy, P.K.2
  • 91
    • 77953619214 scopus 로고    scopus 로고
    • Physiological modeling to understand the impact of enzymes and transporters on drug and metabolite data and bioavailability estimates
    • Sun H, Pang KS,. Physiological modeling to understand the impact of enzymes and transporters on drug and metabolite data and bioavailability estimates. Pharm Res 2010; 27: 1237-1254.
    • (2010) Pharm Res , vol.27 , pp. 1237-1254
    • Sun, H.1    Pang, K.S.2
  • 92
    • 0033009998 scopus 로고    scopus 로고
    • Is the role of the small intestine in first-pass metabolism overemphasized?
    • Lin JH, Chiba M, Baillie TA,. Is the role of the small intestine in first-pass metabolism overemphasized? Pharmacol Rev 1999; 51: 135-158.
    • (1999) Pharmacol Rev , vol.51 , pp. 135-158
    • Lin, J.H.1    Chiba, M.2    Baillie, T.A.3
  • 93
    • 0029738490 scopus 로고    scopus 로고
    • First-pass metabolism of midazolam by the human intestine
    • Paine MF, Shen DD, Kunze KL, et al., First-pass metabolism of midazolam by the human intestine. Clin Pharmacol Ther 1996; 60: 14-24.
    • (1996) Clin Pharmacol Ther , vol.60 , pp. 14-24
    • Paine, M.F.1    Shen, D.D.2    Kunze, K.L.3
  • 94
    • 77953765707 scopus 로고    scopus 로고
    • The role of small-intestinal P450 enzymes in protection against systemic exposure of orally administered benzo [a] pyrene
    • Fang C, Zhang Q-Y,. The role of small-intestinal P450 enzymes in protection against systemic exposure of orally administered benzo [a] pyrene. J Pharmacol Exp Ther 2010; 334: 156-163.
    • (2010) J Pharmacol Exp Ther , vol.334 , pp. 156-163
    • Fang, C.1    Zhang, Q.-Y.2
  • 95
    • 34548092793 scopus 로고    scopus 로고
    • Role of small intestinal cytochromes P450 in the bioavailability of oral nifedipine
    • Zhang Q-Y, Kaminsky LS, Dunbar D, Zhang J, Ding X,. Role of small intestinal cytochromes P450 in the bioavailability of oral nifedipine. Drug Metab Dispos 2007; 35: 1617-1623.
    • (2007) Drug Metab Dispos , vol.35 , pp. 1617-1623
    • Zhang, Q.-Y.1    Kaminsky, L.S.2    Dunbar, D.3    Zhang, J.4    Ding, X.5
  • 96
    • 79956122356 scopus 로고    scopus 로고
    • Role of intestinal cytochrome P450 (P450) in modulating the bioavailability of oral lovastatin: Insights from studies on the intestinal epithelium-specific P450 reductase knockout mouse
    • Zhu Y, D'Agostino J, Zhang Q-Y,. Role of intestinal cytochrome P450 (P450) in modulating the bioavailability of oral lovastatin: insights from studies on the intestinal epithelium-specific P450 reductase knockout mouse. Drug Metab Dispos 2011; 39: 939-943.
    • (2011) Drug Metab Dispos , vol.39 , pp. 939-943
    • Zhu, Y.1    D'Agostino, J.2    Zhang, Q.-Y.3
  • 97
    • 36049034219 scopus 로고    scopus 로고
    • Knockout of cytochrome P450 3A yields new mouse models for understanding xenobiotic metabolism
    • van Herwaarden AE, Wagenaar E, van der Kruijssen CM, et al., Knockout of cytochrome P450 3A yields new mouse models for understanding xenobiotic metabolism. J Clin Invest 2007; 117: 3583-3592.
    • (2007) J Clin Invest , vol.117 , pp. 3583-3592
    • Van Herwaarden, A.E.1    Wagenaar, E.2    Van Der Kruijssen, C.M.3
  • 99
    • 44149087132 scopus 로고    scopus 로고
    • Hydroxyitraconazole, formed during intestinal first-pass metabolism of itraconazole, controls the time course of hepatic CYP3A inhibition and the bioavailability of itraconazole in rats
    • Quinney SK, Galinsky RE, Jiyamapa-Serna VA, et al., Hydroxyitraconazole, formed during intestinal first-pass metabolism of itraconazole, controls the time course of hepatic CYP3A inhibition and the bioavailability of itraconazole in rats. Drug Metab Dispos 2008; 36: 1097-1101.
    • (2008) Drug Metab Dispos , vol.36 , pp. 1097-1101
    • Quinney, S.K.1    Galinsky, R.E.2    Jiyamapa-Serna, V.A.3
  • 100
    • 84455194130 scopus 로고    scopus 로고
    • A semiphysiologically based pharmacokinetic modeling approach to predict the dose-exposure relationship of an antiparasitic prodrug/active metabolite pair
    • Yan GZ, Generaux CN, Yoon M, et al., A semiphysiologically based pharmacokinetic modeling approach to predict the dose-exposure relationship of an antiparasitic prodrug/active metabolite pair. Drug Metab Dispos 2012; 40: 6-17.
    • (2012) Drug Metab Dispos , vol.40 , pp. 6-17
    • Yan, G.Z.1    Generaux, C.N.2    Yoon, M.3
  • 101
    • 61449095680 scopus 로고    scopus 로고
    • An intestinal epithelium-specific cytochrome P450 (P450) reductase-knockout mouse model: Direct evidence for a role of intestinal p450s in first-pass clearance of oral nifedipine
    • Zhang Q-Y, Fang C, Zhang J, Dunbar D, Kaminsky L, Ding X,. An intestinal epithelium-specific cytochrome P450 (P450) reductase-knockout mouse model: direct evidence for a role of intestinal p450s in first-pass clearance of oral nifedipine. Drug Metab Dispos 2009; 37: 651-657.
    • (2009) Drug Metab Dispos , vol.37 , pp. 651-657
    • Zhang, Q.-Y.1    Fang, C.2    Zhang, J.3    Dunbar, D.4    Kaminsky, L.5    Ding, X.6
  • 102
    • 78951482203 scopus 로고    scopus 로고
    • Applications of physiologically based pharmacokinetic (PBPK) modeling and simulation during regulatory review
    • Zhao P, Zhang L, Grillo JA, et al., Applications of physiologically based pharmacokinetic (PBPK) modeling and simulation during regulatory review. Clin Pharmacol Ther 2011; 89: 259-267.
    • (2011) Clin Pharmacol Ther , vol.89 , pp. 259-267
    • Zhao, P.1    Zhang, L.2    Grillo, J.A.3
  • 103
    • 84862776856 scopus 로고    scopus 로고
    • Predicting drug interaction potential with a physiologically based pharmacokinetic model: A case study of telithromycin, a time-dependent CYP3A inhibitor
    • Vieira ML, Zhao P, Berglund EG, et al., Predicting drug interaction potential with a physiologically based pharmacokinetic model: a case study of telithromycin, a time-dependent CYP3A inhibitor. Clin Pharmacol Ther 2012; 91: 700-708.
    • (2012) Clin Pharmacol Ther , vol.91 , pp. 700-708
    • Vieira, M.L.1    Zhao, P.2    Berglund, E.G.3
  • 104
    • 52949140158 scopus 로고    scopus 로고
    • Formed and preformed metabolites: Facts and comparisons
    • Pang KS, Morris ME, Sun H,. Formed and preformed metabolites: facts and comparisons. J Pharm Pharmacol 2008; 60: 1247-1275.
    • (2008) J Pharm Pharmacol , vol.60 , pp. 1247-1275
    • Pang, K.S.1    Morris, M.E.2    Sun, H.3
  • 105
    • 79951500920 scopus 로고    scopus 로고
    • PBPK modeling of intestinal and liver enzymes and transporters in drug absorption and sequential metabolism
    • Fan J, Chen S, Chow EC, Pang KS,. PBPK modeling of intestinal and liver enzymes and transporters in drug absorption and sequential metabolism. Curr Drug Metab 2010; 11: 743-761.
    • (2010) Curr Drug Metab , vol.11 , pp. 743-761
    • Fan, J.1    Chen, S.2    Chow, E.C.3    Pang, K.S.4
  • 106
    • 0028847788 scopus 로고
    • The erythromycin breath test predicts the clearance of midazolam
    • Lown KS, Thummel KE, Benedict PE, et al., The erythromycin breath test predicts the clearance of midazolam. Clin Pharmacol Ther 1995; 57: 16-24.
    • (1995) Clin Pharmacol Ther , vol.57 , pp. 16-24
    • Lown, K.S.1    Thummel, K.E.2    Benedict, P.E.3
  • 107
    • 0029738490 scopus 로고    scopus 로고
    • First-pass metabolism of midazolam by the human intestine
    • Paine MF, Shen DD, Kunze KL, et al., First-pass metabolism of midazolam by the human intestine. Clin Pharmacol Ther 1996; 60: 14-24.
    • (1996) Clin Pharmacol Ther , vol.60 , pp. 14-24
    • Paine, M.F.1    Shen, D.D.2    Kunze, K.L.3
  • 108
    • 0030015297 scopus 로고    scopus 로고
    • Oral first-pass elimination of midazolam involves both gastrointestinal and hepatic CYP3A-mediated metabolism
    • Thummel KE, O'Shea D, Paine MF, et al., Oral first-pass elimination of midazolam involves both gastrointestinal and hepatic CYP3A-mediated metabolism. Clin Pharmacol Ther 1996; 59: 491-502.
    • (1996) Clin Pharmacol Ther , vol.59 , pp. 491-502
    • Thummel, K.E.1    O'Shea, D.2    Paine, M.F.3
  • 109
    • 0034003625 scopus 로고    scopus 로고
    • Route-dependent metabolism of morphine in the vascularly perfused rat small intestine preparation
    • Doherty MM, Pang KS,. Route-dependent metabolism of morphine in the vascularly perfused rat small intestine preparation. Pharm Res 2000; 17: 291-298.
    • (2000) Pharm Res , vol.17 , pp. 291-298
    • Doherty, M.M.1    Pang, K.S.2
  • 110
    • 0032754284 scopus 로고    scopus 로고
    • The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin
    • Greiner B, Eichelbaum M, Fritz P, et al., The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J Clin Invest 1999; 104: 147-153.
    • (1999) J Clin Invest , vol.104 , pp. 147-153
    • Greiner, B.1    Eichelbaum, M.2    Fritz, P.3
  • 112
    • 0033984905 scopus 로고    scopus 로고
    • A new physiologically based, segregated-flow model to explain route-dependent intestinal metabolism
    • Cong D, Doherty M, Pang KS,. A new physiologically based, segregated-flow model to explain route-dependent intestinal metabolism. Drug Metab Dispos 2000; 28: 224-235.
    • (2000) Drug Metab Dispos , vol.28 , pp. 224-235
    • Cong, D.1    Doherty, M.2    Pang, K.S.3
  • 113
    • 80052002510 scopus 로고    scopus 로고
    • Physiologically based pharmacokinetic modeling of intestinal first-pass metabolism of CYP3A substrates with high intestinal extraction
    • Gertz M, Houston JB, Galetin A,. Physiologically based pharmacokinetic modeling of intestinal first-pass metabolism of CYP3A substrates with high intestinal extraction. Drug Metab Dispos 2011; 39: 1633-1642.
    • (2011) Drug Metab Dispos , vol.39 , pp. 1633-1642
    • Gertz, M.1    Houston, J.B.2    Galetin, A.3
  • 115
    • 0021798973 scopus 로고
    • Hepatic elimination - Dispersion model
    • Roberts MS, Rowland M,. Hepatic elimination-dispersion model. J Pharm Sci 1985; 74: 585-587.
    • (1985) J Pharm Sci , vol.74 , pp. 585-587
    • Roberts, M.S.1    Rowland, M.2
  • 116
    • 77953619214 scopus 로고    scopus 로고
    • Physiological modeling to understand the impact of enzymes and transporters on drug and metabolite data and bioavailability estimates
    • Sun H, Pang KS,. Physiological modeling to understand the impact of enzymes and transporters on drug and metabolite data and bioavailability estimates. Pharm Res 2010; 27: 1237-1254.
    • (2010) Pharm Res , vol.27 , pp. 1237-1254
    • Sun, H.1    Pang, K.S.2
  • 118
    • 79956223783 scopus 로고    scopus 로고
    • 3 up-regulates P-glycoprotein via the vitamin D receptor and not farnesoid X receptor in both fxr (-/-) and fxr (+/+) mice and increased renal and brain efflux of digoxin in mice in vivo
    • 3 up-regulates P-glycoprotein via the vitamin D receptor and not farnesoid X receptor in both fxr (-/-) and fxr (+/+) mice and increased renal and brain efflux of digoxin in mice in vivo. J Pharmacol Exp Ther 2011; 337: 846-859.
    • (2011) J Pharmacol Exp Ther , vol.337 , pp. 846-859
    • Chow, E.C.1    Durk, M.R.2    Cummins, C.L.3    Pang, K.S.4
  • 119
    • 77949300138 scopus 로고    scopus 로고
    • Theoretical considerations on quantitative prediction of drug-drug interactions
    • Hisaka A, Ohno Y, Yamamoto T, Suzuki H,. Theoretical considerations on quantitative prediction of drug-drug interactions. Drug Metab Pharmacokinet 2010; 25: 48-61.
    • (2010) Drug Metab Pharmacokinet , vol.25 , pp. 48-61
    • Hisaka, A.1    Ohno, Y.2    Yamamoto, T.3    Suzuki, H.4
  • 120
    • 84888583720 scopus 로고    scopus 로고
    • Drug metabolites as cytochrome P450 inhibitors: A retrospective analysis and proposed algorithm for evaluation of the pharmacokinetic interaction potential of metabolites in drug discovery and development
    • Callegari E, Kalgutkar AS, Leung L, Obach RS, Plowchalk DR, Tse S,. Drug metabolites as cytochrome P450 inhibitors: a retrospective analysis and proposed algorithm for evaluation of the pharmacokinetic interaction potential of metabolites in drug discovery and development. Drug Metab Dispos 2013; 41: 2047-2055.
    • (2013) Drug Metab Dispos , vol.41 , pp. 2047-2055
    • Callegari, E.1    Kalgutkar, A.S.2    Leung, L.3    Obach, R.S.4    Plowchalk, D.R.5    Tse, S.6
  • 121
    • 84874038558 scopus 로고    scopus 로고
    • A perspective on the contribution of metabolites to drug-drug interaction potential: The need to consider both circulating levels and inhibition potency
    • Yu H, Tweedie D,. A perspective on the contribution of metabolites to drug-drug interaction potential: the need to consider both circulating levels and inhibition potency. Drug Metab Dispos 2013; 41: 536-540.
    • (2013) Drug Metab Dispos , vol.41 , pp. 536-540
    • Yu, H.1    Tweedie, D.2
  • 122
    • 62249151758 scopus 로고    scopus 로고
    • Qualitative analysis of the role of metabolites in inhibitory drug-drug interactions: Literature evaluation based on the metabolism and transport drug interaction database
    • Isoherranen N, Hachad H, Yeung CK, Levy RH,. Qualitative analysis of the role of metabolites in inhibitory drug-drug interactions: literature evaluation based on the metabolism and transport drug interaction database. Chem Res Toxicol 2009; 22: 294-298.
    • (2009) Chem Res Toxicol , vol.22 , pp. 294-298
    • Isoherranen, N.1    Hachad, H.2    Yeung, C.K.3    Levy, R.H.4
  • 123
    • 78650513090 scopus 로고    scopus 로고
    • Are circulating metabolites important in drug-drug interactions?: Quantitative analysis of risk prediction and inhibitory potency
    • Yeung CK, Fujioka Y, Hachad H, Levy RH, Isoherranen N,. Are circulating metabolites important in drug-drug interactions?: quantitative analysis of risk prediction and inhibitory potency. Clin Pharmacol Ther 2011; 89: 105-113.
    • (2011) Clin Pharmacol Ther , vol.89 , pp. 105-113
    • Yeung, C.K.1    Fujioka, Y.2    Hachad, H.3    Levy, R.H.4    Isoherranen, N.5
  • 125
    • 37549070318 scopus 로고    scopus 로고
    • Contribution of itraconazole metabolites to inhibition of CYP3A4 in vivo
    • Templeton I, Thummel KE, Kharasch ED, et al., Contribution of itraconazole metabolites to inhibition of CYP3A4 in vivo. Clin Pharmacol Ther 2008; 83: 77-85.
    • (2008) Clin Pharmacol Ther , vol.83 , pp. 77-85
    • Templeton, I.1    Thummel, K.E.2    Kharasch, E.D.3
  • 126
    • 77957019850 scopus 로고    scopus 로고
    • Accurate prediction of dose-dependent CYP3A4 inhibition by itraconazole and its metabolites from in vitro inhibition data
    • Templeton I, Peng C, Thummel K, Davis C, Kunze K, Isoherranen N,. Accurate prediction of dose-dependent CYP3A4 inhibition by itraconazole and its metabolites from in vitro inhibition data. Clin Pharmacol Ther 2010; 88: 499-505.
    • (2010) Clin Pharmacol Ther , vol.88 , pp. 499-505
    • Templeton, I.1    Peng, C.2    Thummel, K.3    Davis, C.4    Kunze, K.5    Isoherranen, N.6
  • 127
    • 73849120796 scopus 로고    scopus 로고
    • The role of metabolites in predicting drug-drug interactions: Focus on irreversible P450 inhibition
    • VandenBrink BM, Isoherranen N,. The role of metabolites in predicting drug-drug interactions: focus on irreversible P450 inhibition. Curr Opin Drug Discov Devel 2010; 13: 66.
    • (2010) Curr Opin Drug Discov Devel , vol.13 , pp. 66
    • Vandenbrink, B.M.1    Isoherranen, N.2
  • 128
    • 0037369622 scopus 로고    scopus 로고
    • Apparent mechanism-based inhibition of human CYP2D6 in vitro by paroxetine: Comparison with fluoxetine and quinidine
    • Bertelsen KM, Venkatakrishnan K, Von Moltke LL, Obach RS, Greenblatt DJ,. Apparent mechanism-based inhibition of human CYP2D6 in vitro by paroxetine: comparison with fluoxetine and quinidine. Drug Metab Dispos 2003; 31: 289-293.
    • (2003) Drug Metab Dispos , vol.31 , pp. 289-293
    • Bertelsen, K.M.1    Venkatakrishnan, K.2    Von Moltke, L.L.3    Obach, R.S.4    Greenblatt, D.J.5
  • 129
    • 77952312729 scopus 로고    scopus 로고
    • Sequential metabolism of secondary alkyl amines to metabolic-intermediate complexes: Opposing roles for the secondary hydroxylamine and primary amine metabolites of desipramine,(s)-fluoxetine, and N-desmethyldiltiazem
    • Hanson KL, VandenBrink BM, Babu KN, Allen KE, Nelson WL, Kunze KL,. Sequential metabolism of secondary alkyl amines to metabolic-intermediate complexes: opposing roles for the secondary hydroxylamine and primary amine metabolites of desipramine,(s)-fluoxetine, and N-desmethyldiltiazem. Drug Metab Dispos 2010; 38: 963-972.
    • (2010) Drug Metab Dispos , vol.38 , pp. 963-972
    • Hanson, K.L.1    Vandenbrink, B.M.2    Babu, K.N.3    Allen, K.E.4    Nelson, W.L.5    Kunze, K.L.6
  • 130
    • 58149464706 scopus 로고    scopus 로고
    • Prediction of the effect of erythromycin, diltiazem, and their metabolites, alone and in combination, on CYP3A4 inhibition
    • Zhang X, Jones DR, Hall SD,. Prediction of the effect of erythromycin, diltiazem, and their metabolites, alone and in combination, on CYP3A4 inhibition. Drug Metab Dispos 2009; 37: 150-160.
    • (2009) Drug Metab Dispos , vol.37 , pp. 150-160
    • Zhang, X.1    Jones, D.R.2    Hall, S.D.3
  • 131
    • 84879068823 scopus 로고    scopus 로고
    • Inhibition of CYP2C19 and CYP3A4 by omeprazole metabolites and their contribution to drug-drug interactions
    • Shirasaka Y, Sager JE, Lutz JD, Davis C, Isoherranen N,. Inhibition of CYP2C19 and CYP3A4 by omeprazole metabolites and their contribution to drug-drug interactions. Drug Metab Dispos 2013; 41: 1414-1424.
    • (2013) Drug Metab Dispos , vol.41 , pp. 1414-1424
    • Shirasaka, Y.1    Sager, J.E.2    Lutz, J.D.3    Davis, C.4    Isoherranen, N.5
  • 132
    • 84888580865 scopus 로고    scopus 로고
    • Stereoselective inhibition of CYP2C19 and CYP3A4 by fluoxetine and its metabolite: Implications for risk assessment of multiple time-dependent inhibitor systems
    • Lutz JD, VandenBrink BM, Babu KN, Nelson WL, Kunze KL, Isoherranen N,. Stereoselective inhibition of CYP2C19 and CYP3A4 by fluoxetine and its metabolite: implications for risk assessment of multiple time-dependent inhibitor systems. Drug Metab Dispos 2013; 41: 2056-2065.
    • (2013) Drug Metab Dispos , vol.41 , pp. 2056-2065
    • Lutz, J.D.1    Vandenbrink, B.M.2    Babu, K.N.3    Nelson, W.L.4    Kunze, K.L.5    Isoherranen, N.6
  • 133
    • 84921630257 scopus 로고    scopus 로고
    • Glucuronidation converts clopidogrel to a strong time-dependent inhibitor of CYP2C8: A phase II metabolite as a perpetrator of drug-drug interactions
    • Tornio A, Filppula AM, Kailari O, et al., Glucuronidation converts clopidogrel to a strong time-dependent inhibitor of CYP2C8: a phase II metabolite as a perpetrator of drug-drug interactions. Clin Pharmacol Ther 2014; 96: 498-507.
    • (2014) Clin Pharmacol Ther , vol.96 , pp. 498-507
    • Tornio, A.1    Filppula, A.M.2    Kailari, O.3
  • 134
    • 84862776909 scopus 로고    scopus 로고
    • Utility of a physiologically-based pharmacokinetic (PBPK) modeling approach to quantitatively predict a complex drug-drug-disease interaction scenario for rivaroxaban during the drug review process: Implications for clinical practice
    • Grillo JA, Zhao P, Bullock J, et al., Utility of a physiologically-based pharmacokinetic (PBPK) modeling approach to quantitatively predict a complex drug-drug-disease interaction scenario for rivaroxaban during the drug review process: implications for clinical practice. Biopharm Drug Dispos 2012; 33: 99-110.
    • (2012) Biopharm Drug Dispos , vol.33 , pp. 99-110
    • Grillo, J.A.1    Zhao, P.2    Bullock, J.3
  • 135
    • 84964314710 scopus 로고    scopus 로고
    • An open-label study to estimate the effect of steady-state erythromycin on the pharmacokinetics, pharmacodynamics, and safety of a single dose of rivaroxaban in subjects with renal impairment and normal renal function
    • Moore KT, Vaidyanathan S, Natarajan J, Ariyawansa J, Haskell L, Turner KC,. An open-label study to estimate the effect of steady-state erythromycin on the pharmacokinetics, pharmacodynamics, and safety of a single dose of rivaroxaban in subjects with renal impairment and normal renal function. J Clin Pharmacol 2014; 54: 1407-1420.
    • (2014) J Clin Pharmacol , vol.54 , pp. 1407-1420
    • Moore, K.T.1    Vaidyanathan, S.2    Natarajan, J.3    Ariyawansa, J.4    Haskell, L.5    Turner, K.C.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.