Skip to main content
Erschienen in: Clinical Pharmacokinetics 8/2014

01.08.2014 | Current Opinion

Prediction of Pharmacokinetics and Drug–Drug Interactions When Hepatic Transporters are Involved

verfasst von: Rui Li, Hugh A. Barton, Manthena V. Varma

Erschienen in: Clinical Pharmacokinetics | Ausgabe 8/2014

Einloggen, um Zugang zu erhalten

Abstract

Hepatobiliary transport mechanisms have been identified to play a significant role in determining the systemic clearance for a number of widely prescribed drugs and an increasing number of new molecular entities (NMEs). While determining the pharmacokinetics, drug transporters also regulate the target tissue exposure and play a key role in regulating the pharmacological and/or toxicological responses. Consequently, it is of great relevance in drug discovery and development to assess hepatic transporter activity in regard to pharmacokinetic and dose predictions and to evaluate pharmacokinetic variability associated with drug–drug interactions (DDIs) and genetic variants. Mechanistic predictions utilizing physiological-based pharmacokinetic modeling are increasingly used to evaluate transporter contribution and delineate the transporter–enzyme interplay on the basis of hypothesis-driven functional in vitro findings. Significant strides were made in the development of in vitro techniques to facilitate characterization of hepatobiliary transport. However, challenges exist in the quantitative in vitro–in vivo extrapolation of transporter kinetics due to the lack of information on absolute abundance of the transporter in both in vitro and in vivo situations, and/or differential function in the holistic in vitro reagents such as suspended and plated hepatocytes systems, and lack of complete mechanistic understanding of liver model structure. On the other hand, models to predict transporter-mediated DDIs range from basic models to mechanistic static and dynamic models. While basic models provide conservative estimates and are useful upfront in avoiding false negative predictions, mechanistic models integrate multiple victim and perpetrator drugs parameters and are expected to provide quantitative predictions. The aim of this paper is to review the current state of the model-based approaches to predict clinical pharmacokinetics and DDIs of drugs or NMEs that are substrates of hepatic transporters.
Literatur
1.
Zurück zum Zitat Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci. 2006;27(5):425–46.PubMed Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci. 2006;27(5):425–46.PubMed
2.
Zurück zum Zitat Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92(16):1295–302.PubMed Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92(16):1295–302.PubMed
3.
Zurück zum Zitat Hagenbuch B, Meier P. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta. 2003;1609(1):1–18.PubMed Hagenbuch B, Meier P. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta. 2003;1609(1):1–18.PubMed
4.
Zurück zum Zitat Hagenbuch B, Meier PJ. Organic anion transporting polypeptides of the OATP/SLC21 family: phylogenetic classification as OATP/SLCO superfamily, new nomenclature and molecular/functional properties. Pflugers Arch. 2004;447(5):653–65.PubMed Hagenbuch B, Meier PJ. Organic anion transporting polypeptides of the OATP/SLC21 family: phylogenetic classification as OATP/SLCO superfamily, new nomenclature and molecular/functional properties. Pflugers Arch. 2004;447(5):653–65.PubMed
5.
Zurück zum Zitat Tamai I, Nezu J, Uchino H, Sai Y, Oku A, Shimane M, et al. Molecular identification and characterization of novel members of the human organic anion transporter (OATP) family. Biochem Biophys Res Commun. 2000;273(1):251–60.PubMed Tamai I, Nezu J, Uchino H, Sai Y, Oku A, Shimane M, et al. Molecular identification and characterization of novel members of the human organic anion transporter (OATP) family. Biochem Biophys Res Commun. 2000;273(1):251–60.PubMed
6.
Zurück zum Zitat Fahrmayr C, Fromm MF, Konig J. Hepatic OATP and OCT uptake transporters: their role for drug–drug interactions and pharmacogenetic aspects. Drug Metab Rev. 2010;42(3):380–401.PubMed Fahrmayr C, Fromm MF, Konig J. Hepatic OATP and OCT uptake transporters: their role for drug–drug interactions and pharmacogenetic aspects. Drug Metab Rev. 2010;42(3):380–401.PubMed
7.
Zurück zum Zitat 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. 2011;42(1):28–45.PubMed 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. 2011;42(1):28–45.PubMed
8.
Zurück zum Zitat Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36.PubMed Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36.PubMed
9.
Zurück zum Zitat 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(1):157–81.PubMed 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(1):157–81.PubMed
10.
Zurück zum Zitat 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(6):1053–64.PubMed 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(6):1053–64.PubMed
11.
Zurück zum Zitat Kock K, Brouwer KL. A perspective on efflux transport proteins in the liver. Clin Pharmacol Ther. 2012;92(5):599–612.PubMedCentralPubMed Kock K, Brouwer KL. A perspective on efflux transport proteins in the liver. Clin Pharmacol Ther. 2012;92(5):599–612.PubMedCentralPubMed
12.
Zurück zum Zitat Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol Rev. 2010;62(1):1–96.PubMedCentralPubMed Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol Rev. 2010;62(1):1–96.PubMedCentralPubMed
13.
Zurück zum Zitat Pfeifer ND, Hardwick RN, Brouwer KL. Role of hepatic efflux transporters in regulating systemic and hepatocyte exposure to xenobiotics. Annu Rev Pharmacol Toxicol. 2014;54:509–35.PubMed Pfeifer ND, Hardwick RN, Brouwer KL. Role of hepatic efflux transporters in regulating systemic and hepatocyte exposure to xenobiotics. Annu Rev Pharmacol Toxicol. 2014;54:509–35.PubMed
14.
Zurück zum Zitat Pfeifer ND, Yang K, Brouwer KL. Hepatic basolateral efflux contributes significantly to rosuvastatin disposition I: characterization of basolateral versus biliary clearance using a novel protocol in sandwich-cultured hepatocytes. J Pharmacol Exp Ther. 2013;347(3):727–36.PubMed Pfeifer ND, Yang K, Brouwer KL. Hepatic basolateral efflux contributes significantly to rosuvastatin disposition I: characterization of basolateral versus biliary clearance using a novel protocol in sandwich-cultured hepatocytes. J Pharmacol Exp Ther. 2013;347(3):727–36.PubMed
15.
Zurück zum Zitat Hagenbuch B, Gui C. Xenobiotic transporters of the human organic anion transporting polypeptides (OATP) family. Xenobiotica. 2008;38(7–8):778–801.PubMed Hagenbuch B, Gui C. Xenobiotic transporters of the human organic anion transporting polypeptides (OATP) family. Xenobiotica. 2008;38(7–8):778–801.PubMed
16.
Zurück zum Zitat Varma MV, Rotter CJ, Chupka J, Whalen KM, Duignan DB, Feng B, et al. pH-sensitive interaction of HMG-CoA reductase inhibitors (statins) with organic anion transporting polypeptide 2B1. Mol Pharm. 2011;8(4):1303–13.PubMed Varma MV, Rotter CJ, Chupka J, Whalen KM, Duignan DB, Feng B, et al. pH-sensitive interaction of HMG-CoA reductase inhibitors (statins) with organic anion transporting polypeptide 2B1. Mol Pharm. 2011;8(4):1303–13.PubMed
17.
Zurück zum Zitat Kobayashi D, Nozawa T, Imai K, Nezu J, Tsuji A, Tamai I. Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane. J Pharmacol Exp Ther. 2003;306(2):703–8.PubMed Kobayashi D, Nozawa T, Imai K, Nezu J, Tsuji A, Tamai I. Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane. J Pharmacol Exp Ther. 2003;306(2):703–8.PubMed
18.
Zurück zum Zitat Leuthold S, Hagenbuch B, Mohebbi N, Wagner CA, Meier PJ, Stieger B. Mechanisms of pH-gradient driven transport mediated by organic anion polypeptide transporters. Am J Physiol Cell Physiol. 2009;296(3):C570–82.PubMed Leuthold S, Hagenbuch B, Mohebbi N, Wagner CA, Meier PJ, Stieger B. Mechanisms of pH-gradient driven transport mediated by organic anion polypeptide transporters. Am J Physiol Cell Physiol. 2009;296(3):C570–82.PubMed
19.
Zurück zum Zitat 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 Dispos. 2012;40(8):1527–37.PubMed 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 Dispos. 2012;40(8):1527–37.PubMed
20.
Zurück zum Zitat 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(4):459–72.PubMed 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(4):459–72.PubMed
21.
Zurück zum Zitat 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(1):71–105.PubMed 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(1):71–105.PubMed
22.
Zurück zum Zitat Group SC, Link E, Parish S, Armitage J, Bowman L, Heath S. SLCO1B1 variants and statin-induced myopathy–a genomewide study. N Engl J Med. 2008;359(8):789–99.PubMed Group SC, Link E, Parish S, Armitage J, Bowman L, Heath S. SLCO1B1 variants and statin-induced myopathy–a genomewide study. N Engl J Med. 2008;359(8):789–99.PubMed
23.
24.
Zurück zum Zitat Treiber A, Schneiter R, Häusler S, Stieger B. Bosentan is a substrate of human OATP1B1 and OATP1B3: inhibition of hepatic uptake as the common mechanism of its interactions with cyclosporin A, rifampicin, and sildenafil. Drug Metab Dispos. 2007;35(8):1400–7.PubMed Treiber A, Schneiter R, Häusler S, Stieger B. Bosentan is a substrate of human OATP1B1 and OATP1B3: inhibition of hepatic uptake as the common mechanism of its interactions with cyclosporin A, rifampicin, and sildenafil. Drug Metab Dispos. 2007;35(8):1400–7.PubMed
25.
Zurück zum Zitat 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 Dispos. 2006;34(7):1247–54.PubMed 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 Dispos. 2006;34(7):1247–54.PubMed
26.
Zurück zum Zitat EMA. Guideline on the investigation of drug interactions. Committee for Human Medicinal Products (CHMP). London: EMA; 2012. (CPMP/EWP/560/95/Rev). EMA. Guideline on the investigation of drug interactions. Committee for Human Medicinal Products (CHMP). London: EMA; 2012. (CPMP/EWP/560/95/Rev).
27.
Zurück zum Zitat US FDA. Guidance for industry. Drug interaction studies—study design, data analysis, implications for dosing, and labeling recommendations. Center for Drug Evaluation and Research (CDER). Silver Spring: FDA; 2012. US FDA. Guidance for industry. Drug interaction studies—study design, data analysis, implications for dosing, and labeling recommendations. Center for Drug Evaluation and Research (CDER). Silver Spring: FDA; 2012.
28.
Zurück zum Zitat 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(1):45–78.PubMed 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(1):45–78.PubMed
29.
Zurück zum Zitat Ogasawara K, Terada T, Katsura T, Hatano E, Ikai I, Yamaoka Y, et al. Hepatitis C virus-related cirrhosis is a major determinant of the expression levels of hepatic drug transporters. Drug Metab Pharmacokinet. 2010;25(2):190–9.PubMed Ogasawara K, Terada T, Katsura T, Hatano E, Ikai I, Yamaoka Y, et al. Hepatitis C virus-related cirrhosis is a major determinant of the expression levels of hepatic drug transporters. Drug Metab Pharmacokinet. 2010;25(2):190–9.PubMed
30.
Zurück zum Zitat Farmer JA. Learning from the cerivastatin experience. Lancet. 2001;358(9291):1383–5.PubMed Farmer JA. Learning from the cerivastatin experience. Lancet. 2001;358(9291):1383–5.PubMed
31.
Zurück zum Zitat Lau YY, Huang Y, Frassetto L, Benet LZ. Effect of OATP1B transporter inhibition on the pharmacokinetics of atorvastatin in healthy volunteers. Clin Pharmacol Ther. 2007;81(2):194–204.PubMed Lau YY, Huang Y, Frassetto L, Benet LZ. Effect of OATP1B transporter inhibition on the pharmacokinetics of atorvastatin in healthy volunteers. Clin Pharmacol Ther. 2007;81(2):194–204.PubMed
32.
Zurück zum Zitat Regazzi MB, Iacona I, Campana C, Raddato V, Lesi C, Perani G, et al. Altered disposition of pravastatin following concomitant drug therapy with cyclosporin A in transplant recipients. Transplant Proc. 1993;25(4):2732–4.PubMed Regazzi MB, Iacona I, Campana C, Raddato V, Lesi C, Perani G, et al. Altered disposition of pravastatin following concomitant drug therapy with cyclosporin A in transplant recipients. Transplant Proc. 1993;25(4):2732–4.PubMed
33.
Zurück zum Zitat Schneck DW, Birmingham BK, Zalikowski JA, Mitchell PD, Wang Y, Martin PD, et al. The effect of gemfibrozil on the pharmacokinetics of rosuvastatin. Clin Pharmacol Ther. 2004;75(5):455–63.PubMed Schneck DW, Birmingham BK, Zalikowski JA, Mitchell PD, Wang Y, Martin PD, et al. The effect of gemfibrozil on the pharmacokinetics of rosuvastatin. Clin Pharmacol Ther. 2004;75(5):455–63.PubMed
34.
Zurück zum Zitat 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(4):1188–99.PubMed 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(4):1188–99.PubMed
35.
Zurück zum Zitat 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(4):575–81.PubMed 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(4):575–81.PubMed
36.
Zurück zum Zitat 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(1):37–51.PubMed 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(1):37–51.PubMed
37.
Zurück zum Zitat Niemi M, Schaeffeler E, Lang T, Fromm MF, Neuvonen M, Kyrklund C, et al. High plasma pravastatin concentrations are associated with single nucleotide polymorphisms and haplotypes of organic anion transporting polypeptide-C (OATP-C, SLCO1B1). Pharmacogenetics. 2004;14(7):429–40.PubMed Niemi M, Schaeffeler E, Lang T, Fromm MF, Neuvonen M, Kyrklund C, et al. High plasma pravastatin concentrations are associated with single nucleotide polymorphisms and haplotypes of organic anion transporting polypeptide-C (OATP-C, SLCO1B1). Pharmacogenetics. 2004;14(7):429–40.PubMed
38.
Zurück zum Zitat 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(6):723–43.PubMed 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(6):723–43.PubMed
39.
Zurück zum Zitat Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J. 2004;5(1):6–13. Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J. 2004;5(1):6–13.
40.
Zurück zum Zitat International Warfarin Pharmacogenetics Consortium, Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, et al. Estimation of the warfarin dose with clinical and pharmacogenetic data. N Engl J Med. 2009;360:753–64.PubMed International Warfarin Pharmacogenetics Consortium, Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, et al. Estimation of the warfarin dose with clinical and pharmacogenetic data. N Engl J Med. 2009;360:753–64.PubMed
41.
Zurück zum Zitat 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(7):703–29.PubMed 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(7):703–29.PubMed
42.
Zurück zum Zitat 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(5):303–9.PubMed 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(5):303–9.PubMed
43.
Zurück zum Zitat Nishizato Y, Ieiri I, Suzuki H, Kimura M, Kawabata K, Hirota T, et al. Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: consequences for pravastatin pharmacokinetics. Clin Pharmacol Ther. 2003;73(6):554–65.PubMed Nishizato Y, Ieiri I, Suzuki H, Kimura M, Kawabata K, Hirota T, et al. Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: consequences for pravastatin pharmacokinetics. Clin Pharmacol Ther. 2003;73(6):554–65.PubMed
44.
Zurück zum Zitat Deo AK, Prasad B, Balogh L, Lai Y, Unadkat JD. Interindividual variability in hepatic expression of the multidrug resistance-associated protein 2 (MRP2/ABCC2): quantification by liquid chromatography/tandem mass spectrometry. Drug Metab Dispos. 2012;40(5):852–5.PubMedCentralPubMed Deo AK, Prasad B, Balogh L, Lai Y, Unadkat JD. Interindividual variability in hepatic expression of the multidrug resistance-associated protein 2 (MRP2/ABCC2): quantification by liquid chromatography/tandem mass spectrometry. Drug Metab Dispos. 2012;40(5):852–5.PubMedCentralPubMed
45.
Zurück zum Zitat Prasad B, Evers R, Gupta A, Hop CE, Salphati L, Shukla S, et al. Interindividual variability in hepatic organic anion-transporting polypeptides and P-glycoprotein (ABCB1) protein expression: quantification by liquid chromatography tandem mass spectroscopy and influence of genotype, age, and sex. Drug Metab Dispos. 2014;42(1):78–88.PubMedCentralPubMed Prasad B, Evers R, Gupta A, Hop CE, Salphati L, Shukla S, et al. Interindividual variability in hepatic organic anion-transporting polypeptides and P-glycoprotein (ABCB1) protein expression: quantification by liquid chromatography tandem mass spectroscopy and influence of genotype, age, and sex. Drug Metab Dispos. 2014;42(1):78–88.PubMedCentralPubMed
46.
Zurück zum Zitat Prasad B, Lai Y, Lin Y, Unadkat JD. Interindividual variability in the hepatic expression of the human breast cancer resistance protein (BCRP/ABCG2): effect of age, sex, and genotype. J Pharm Sci. 2013;102(3):787–93.PubMed Prasad B, Lai Y, Lin Y, Unadkat JD. Interindividual variability in the hepatic expression of the human breast cancer resistance protein (BCRP/ABCG2): effect of age, sex, and genotype. J Pharm Sci. 2013;102(3):787–93.PubMed
47.
Zurück zum Zitat Cheng Q, Aleksunes LM, Manautou JE, Cherrington NJ, Scheffer GL, Yamasaki H, et al. Drug-metabolizing enzyme and transporter expression in a mouse model of diabetes and obesity. Mol Pharm. 2008;5(1):77–91.PubMed Cheng Q, Aleksunes LM, Manautou JE, Cherrington NJ, Scheffer GL, Yamasaki H, et al. Drug-metabolizing enzyme and transporter expression in a mouse model of diabetes and obesity. Mol Pharm. 2008;5(1):77–91.PubMed
48.
Zurück zum Zitat Mahmood I, Balian JD. Interspecies scaling: predicting pharmacokinetic parameters of antiepileptic drugs in humans from animals with special emphasis on clearance. J Pharm Sci. 1996;85(4):411–4.PubMed Mahmood I, Balian JD. Interspecies scaling: predicting pharmacokinetic parameters of antiepileptic drugs in humans from animals with special emphasis on clearance. J Pharm Sci. 1996;85(4):411–4.PubMed
49.
Zurück zum Zitat Kang HE, Lee MG. Approaches for predicting human pharmacokinetics using interspecies pharmacokinetic scaling. Arch Pharm Res. 2011;34(11):1779–88.PubMed Kang HE, Lee MG. Approaches for predicting human pharmacokinetics using interspecies pharmacokinetic scaling. Arch Pharm Res. 2011;34(11):1779–88.PubMed
50.
Zurück zum Zitat Houston JB. Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol. 1994;47(9):1469–79.PubMed Houston JB. Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol. 1994;47(9):1469–79.PubMed
51.
Zurück zum Zitat Ito K, Houston JB. Comparison of the use of liver models for predicting drug clearance using in vitro kinetic data from hepatic microsomes and isolated hepatocytes. Pharm Res. 2004;21(5):785–92.PubMed Ito K, Houston JB. Comparison of the use of liver models for predicting drug clearance using in vitro kinetic data from hepatic microsomes and isolated hepatocytes. Pharm Res. 2004;21(5):785–92.PubMed
52.
Zurück zum Zitat 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(6):625–53.PubMed 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(6):625–53.PubMed
53.
Zurück zum Zitat Wilkinson GR, Shand DG. Commentary: a physiological approach to hepatic drug clearance. Clin Pharmacol Ther. 1975;18(4):377–90.PubMed Wilkinson GR, Shand DG. Commentary: a physiological approach to hepatic drug clearance. Clin Pharmacol Ther. 1975;18(4):377–90.PubMed
54.
Zurück zum Zitat Roberts MS, Rowland M. A dispersion model of hepatic elimination: 1. Formulation of the model and bolus considerations. J Pharmacokinet Biopharm. 1986;14(3):227–60.PubMed Roberts MS, Rowland M. A dispersion model of hepatic elimination: 1. Formulation of the model and bolus considerations. J Pharmacokinet Biopharm. 1986;14(3):227–60.PubMed
55.
Zurück zum Zitat Fagerholm U. Presentation of a modified dispersion model (MDM) for hepatic drug extraction and a new methodology for the prediction of the rate-limiting step in hepatic metabolic clearance. Xenobiotica. 2009;39(1):57–71.PubMed Fagerholm U. Presentation of a modified dispersion model (MDM) for hepatic drug extraction and a new methodology for the prediction of the rate-limiting step in hepatic metabolic clearance. Xenobiotica. 2009;39(1):57–71.PubMed
56.
Zurück zum Zitat 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(5):331–47.PubMed 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(5):331–47.PubMed
57.
Zurück zum Zitat Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73.PubMed Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73.PubMed
58.
Zurück zum Zitat Rowland M. Physiologically-based pharmacokinetic (PBPK) modeling and simulations principles, methods, and applications in the pharmaceutical industry. CPT Pharmacometrics Syst Pharmacol. 2013;2:e55.PubMedCentralPubMed Rowland M. Physiologically-based pharmacokinetic (PBPK) modeling and simulations principles, methods, and applications in the pharmaceutical industry. CPT Pharmacometrics Syst Pharmacol. 2013;2:e55.PubMedCentralPubMed
59.
Zurück zum Zitat 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(7):1237–54.PubMed 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(7):1237–54.PubMed
60.
Zurück zum Zitat 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(10):1766–80.PubMed 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(10):1766–80.PubMed
61.
Zurück zum Zitat Liu L, Pang KS. The roles of transporters and enzymes in hepatic drug processing. Drug Metab Dispos. 2005;33(1):1–9.PubMed Liu L, Pang KS. The roles of transporters and enzymes in hepatic drug processing. Drug Metab Dispos. 2005;33(1):1–9.PubMed
62.
Zurück zum Zitat 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.PubMed 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.PubMed
63.
Zurück zum Zitat Dobson PD, Kell DB. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat Rev Drug Discov. 2008;7(3):205–20.PubMed Dobson PD, Kell DB. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat Rev Drug Discov. 2008;7(3):205–20.PubMed
64.
Zurück zum Zitat 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(2):215–22.PubMed 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(2):215–22.PubMed
65.
Zurück zum Zitat 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(4):179–94.PubMed 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(4):179–94.PubMed
66.
Zurück zum Zitat Menochet 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(9):1744–56.PubMedCentralPubMed Menochet 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(9):1744–56.PubMedCentralPubMed
67.
Zurück zum Zitat 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 Dispos. 2012;40(5):1007–17.PubMed 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 Dispos. 2012;40(5):1007–17.PubMed
68.
Zurück zum Zitat Jamei M, Bajot F, Neuhoff S, Barter Z, Yang J, Rostami-Hodjegan A, 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(1):73–87.PubMedCentralPubMed Jamei M, Bajot F, Neuhoff S, Barter Z, Yang J, Rostami-Hodjegan A, 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(1):73–87.PubMedCentralPubMed
69.
Zurück zum Zitat Simonson SG, Raza A, Martin PD, Mitchell PD, Jarcho JA, Brown CD, et al. Rosuvastatin pharmacokinetics in heart transplant recipients administered an antirejection regimen including cyclosporine. Clin Pharmacol Ther. 2004;76(2):167–77.PubMed Simonson SG, Raza A, Martin PD, Mitchell PD, Jarcho JA, Brown CD, et al. Rosuvastatin pharmacokinetics in heart transplant recipients administered an antirejection regimen including cyclosporine. Clin Pharmacol Ther. 2004;76(2):167–77.PubMed
70.
Zurück zum Zitat 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(7):607–21.PubMed 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(7):607–21.PubMed
71.
Zurück zum Zitat 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(10):2860–73.PubMed 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(10):2860–73.PubMed
72.
Zurück zum Zitat 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(2):652–62.PubMed 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(2):652–62.PubMed
73.
Zurück zum Zitat 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(4):488–96.PubMed 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(4):488–96.PubMed
74.
Zurück zum Zitat Poirier A, Cascais AC, Funk C, Lave T. Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data. J Pharmacokinet Pharmacodyn. 2009;36(6):585–611.PubMed Poirier A, Cascais AC, Funk C, Lave T. Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data. J Pharmacokinet Pharmacodyn. 2009;36(6):585–611.PubMed
75.
Zurück zum Zitat Zamek-Gliszczynski MJ, Lee CA, Poirier A, Bentz J, Chu X, Ellens H, et al. ITC recommendations for transporter kinetic parameter estimation and translational modeling of transport-mediated PK and DDIs in humans. Clin Pharmacol Ther. 2013;94(1):64–79.PubMedCentralPubMed Zamek-Gliszczynski MJ, Lee CA, Poirier A, Bentz J, Chu X, Ellens H, et al. ITC recommendations for transporter kinetic parameter estimation and translational modeling of transport-mediated PK and DDIs in humans. Clin Pharmacol Ther. 2013;94(1):64–79.PubMedCentralPubMed
76.
Zurück zum Zitat Pfeifer ND, Goss SL, Swift B, Ghibellini G, Ivanovic M, Heizer WD, et al. Effect of ritonavir on (99 m) technetium–mebrofenin disposition in humans: a semi-PBPK modeling and in vitro approach to predict transporter-mediated DDIs. CPT Pharmacometrics Syst Pharmacol. 2013;2:e20.PubMedCentralPubMed Pfeifer ND, Goss SL, Swift B, Ghibellini G, Ivanovic M, Heizer WD, et al. Effect of ritonavir on (99 m) technetium–mebrofenin disposition in humans: a semi-PBPK modeling and in vitro approach to predict transporter-mediated DDIs. CPT Pharmacometrics Syst Pharmacol. 2013;2:e20.PubMedCentralPubMed
77.
Zurück zum Zitat Poirier A, Funk C, Scherrmann JM, Lave T. Mechanistic modeling of hepatic transport from cells to whole body: application to napsagatran and fexofenadine. Mol Pharm. 2009;6(6):1716–33.PubMed Poirier A, Funk C, Scherrmann JM, Lave T. Mechanistic modeling of hepatic transport from cells to whole body: application to napsagatran and fexofenadine. Mol Pharm. 2009;6(6):1716–33.PubMed
78.
Zurück zum Zitat 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. Epub 2014 Mar 13. doi:10.1007/s11095-014-1333-3 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. Epub 2014 Mar 13. doi:10.​1007/​s11095-014-1333-3
79.
Zurück zum Zitat Kimoto E, Yoshida K, Balogh LM, Bi YA, Maeda K, El-Kattan A, et al. Characterization of organic anion transporting polypeptide (OATP) expression and its functional contribution to the uptake of substrates in human hepatocytes. Mol Pharm. 2012;9(12):3535–42.PubMed Kimoto E, Yoshida K, Balogh LM, Bi YA, Maeda K, El-Kattan A, et al. Characterization of organic anion transporting polypeptide (OATP) expression and its functional contribution to the uptake of substrates in human hepatocytes. Mol Pharm. 2012;9(12):3535–42.PubMed
80.
Zurück zum Zitat 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(1):139–46.PubMed 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(1):139–46.PubMed
81.
Zurück zum Zitat Kitamura S, Maeda K, Wang Y, Sugiyama Y. Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin. Drug Metab Dispos. 2008;36(10):2014–23.PubMed Kitamura S, Maeda K, Wang Y, Sugiyama Y. Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin. Drug Metab Dispos. 2008;36(10):2014–23.PubMed
82.
Zurück zum Zitat Yamada A, Maeda K, Kamiyama E, Sugiyama D, Kondo T, Shiroyanagi Y, et al. Multiple human isoforms of drug transporters contribute to the hepatic and renal transport of olmesartan, a selective antagonist of the angiotensin II AT1-receptor. Drug Metab Dispos. 2007;35(12):2166–76.PubMed Yamada A, Maeda K, Kamiyama E, Sugiyama D, Kondo T, Shiroyanagi Y, et al. Multiple human isoforms of drug transporters contribute to the hepatic and renal transport of olmesartan, a selective antagonist of the angiotensin II AT1-receptor. Drug Metab Dispos. 2007;35(12):2166–76.PubMed
83.
Zurück zum Zitat Williamson B, Soars AC, Owen A, White P, Riley RJ, Soars MG. Dissecting the relative contribution of OATP1B1-mediated uptake of xenobiotics into human hepatocytes using siRNA. Xenobiotica. 2013;43(10):920–31.PubMed Williamson B, Soars AC, Owen A, White P, Riley RJ, Soars MG. Dissecting the relative contribution of OATP1B1-mediated uptake of xenobiotics into human hepatocytes using siRNA. Xenobiotica. 2013;43(10):920–31.PubMed
84.
Zurück zum Zitat Hatorp V, Oliver S, Su CA. Bioavailability of repaglinide, a novel antidiabetic agent, administered orally in tablet or solution form or intravenously in healthy male volunteers. Int J Clin Pharmacol Ther. 1998;36(12):636–41.PubMed Hatorp V, Oliver S, Su CA. Bioavailability of repaglinide, a novel antidiabetic agent, administered orally in tablet or solution form or intravenously in healthy male volunteers. Int J Clin Pharmacol Ther. 1998;36(12):636–41.PubMed
85.
Zurück zum Zitat Li R, Barton HA, Yates PD, Ghosh A, Wolford AC, Riccardi KA, et al. A “middle-out” approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn. 2014;41(3):197–209.PubMed Li R, Barton HA, Yates PD, Ghosh A, Wolford AC, Riccardi KA, et al. A “middle-out” approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn. 2014;41(3):197–209.PubMed
86.
Zurück zum Zitat Poulin P, Theil FP. A priori prediction of tissue:plasma partition coefficients of drugs to facilitate the use of physiologically-based pharmacokinetic models in drug discovery. J Pharm Sci. 2000;89(1):16–35.PubMed Poulin P, Theil FP. A priori prediction of tissue:plasma partition coefficients of drugs to facilitate the use of physiologically-based pharmacokinetic models in drug discovery. J Pharm Sci. 2000;89(1):16–35.PubMed
87.
Zurück zum Zitat Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94(6):1259–76.PubMed Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94(6):1259–76.PubMed
88.
Zurück zum Zitat Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95(6):1238–57.PubMed Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95(6):1238–57.PubMed
89.
Zurück zum Zitat Gong IY, Kim RB. Impact of genetic variation in OATP transporters to drug disposition and response. Drug Metab Pharmacokinet. 2013;28(1):4–18.PubMed Gong IY, Kim RB. Impact of genetic variation in OATP transporters to drug disposition and response. Drug Metab Pharmacokinet. 2013;28(1):4–18.PubMed
90.
Zurück zum Zitat 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(2):411–26.PubMedCentralPubMed 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(2):411–26.PubMedCentralPubMed
91.
Zurück zum Zitat Karlgren M, Vildhede A, Norinder U, Wisniewski JR, Kimoto E, Lai Y, 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(10):4740–63.PubMedCentralPubMed Karlgren M, Vildhede A, Norinder U, Wisniewski JR, Kimoto E, Lai Y, 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(10):4740–63.PubMedCentralPubMed
92.
Zurück zum Zitat Rowland M, Matin SB. Kinetics of drug–drug interactions. J Pharmacokinet Pharmacodyn. 1973;1(6):553–67. Rowland M, Matin SB. Kinetics of drug–drug interactions. J Pharmacokinet Pharmacodyn. 1973;1(6):553–67.
93.
Zurück zum Zitat Fahmi OA, Hurst S, Plowchalk D, Cook J, Guo F, Youdim K, 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(8):1658–66.PubMed Fahmi OA, Hurst S, Plowchalk D, Cook J, Guo F, Youdim K, 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(8):1658–66.PubMed
94.
Zurück zum Zitat Obach RS, Walsky RL, Venkatakrishnan K, Gaman EA, Houston JB, Tremaine LM. The utility of in vitro cytochrome P450 inhibition data in the prediction of drug–drug interactions. J Pharmacol Exp Ther. 2006;316(1):336–48.PubMed Obach RS, Walsky RL, Venkatakrishnan K, Gaman EA, Houston JB, Tremaine LM. The utility of in vitro cytochrome P450 inhibition data in the prediction of drug–drug interactions. J Pharmacol Exp Ther. 2006;316(1):336–48.PubMed
95.
Zurück zum Zitat Zamek-Gliszczynski MJ, Kalvass JC, Pollack GM, Brouwer KL. Relationship between drug/metabolite exposure and impairment of excretory transport function. Drug Metab Dispos. 2009;37(2):386–90.PubMedCentralPubMed Zamek-Gliszczynski MJ, Kalvass JC, Pollack GM, Brouwer KL. Relationship between drug/metabolite exposure and impairment of excretory transport function. Drug Metab Dispos. 2009;37(2):386–90.PubMedCentralPubMed
96.
Zurück zum Zitat 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(5):1063–74.PubMed 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(5):1063–74.PubMed
97.
Zurück zum Zitat Hu ZY. Disposition pathway-dependent approach for predicting organic anion-transporting polypeptide-mediated drug–drug interactions. Clin Pharmacokinet. 2013;52(6):433–41.PubMed Hu ZY. Disposition pathway-dependent approach for predicting organic anion-transporting polypeptide-mediated drug–drug interactions. Clin Pharmacokinet. 2013;52(6):433–41.PubMed
98.
Zurück zum Zitat 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(6):575–90.PubMed 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(6):575–90.PubMed
99.
Zurück zum Zitat Jamei M, Marciniak S, Feng K, Barnett A, Tucker G, Rostami-Hodjegan A. The Simcyp population-based ADME simulator. Expert Opin Drug Metab Toxicol. 2009;5(2):211–23.PubMed Jamei M, Marciniak S, Feng K, Barnett A, Tucker G, Rostami-Hodjegan A. The Simcyp population-based ADME simulator. Expert Opin Drug Metab Toxicol. 2009;5(2):211–23.PubMed
100.
Zurück zum Zitat 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 Dispos. 2013;41(5):966–74.PubMed 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 Dispos. 2013;41(5):966–74.PubMed
101.
Zurück zum Zitat 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(8):1698–708.PubMed 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(8):1698–708.PubMed
102.
Zurück zum Zitat 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(11):5172–7.PubMedCentralPubMed 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(11):5172–7.PubMedCentralPubMed
103.
Zurück zum Zitat 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(1):72–87.PubMedCentralPubMed 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(1):72–87.PubMedCentralPubMed
104.
Zurück zum Zitat 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(2):140–8.PubMed 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(2):140–8.PubMed
105.
Zurück zum Zitat 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(8):1495–507.PubMed 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(8):1495–507.PubMed
106.
Zurück zum Zitat Gertz M, Cartwright CM, Hobbs MJ, Kenworthy KE, Rowland M, Houston JB, et al. Cyclosporine inhibition of hepatic and intestinal CYP3A4, uptake and efflux transporters: application of PBPK modeling in the assessment of drug–drug interaction potential. Pharm Res. 2013;30(3):761–80.PubMed Gertz M, Cartwright CM, Hobbs MJ, Kenworthy KE, Rowland M, Houston JB, et al. Cyclosporine inhibition of hepatic and intestinal CYP3A4, uptake and efflux transporters: application of PBPK modeling in the assessment of drug–drug interaction potential. Pharm Res. 2013;30(3):761–80.PubMed
107.
Zurück zum Zitat 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(4):736–48.PubMed 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(4):736–48.PubMed
108.
Zurück zum Zitat Bi YA, Qiu X, Rotter CJ, Kimoto E, Piotrowski M, Varma MV, et al. Quantitative assessment of the contribution of sodium-dependent taurocholate co-transporting polypeptide (NTCP) to the hepatic uptake of rosuvastatin, pitavastatin and fluvastatin. Biopharm Drug Dispos. 2013;34(8):452–61.PubMed Bi YA, Qiu X, Rotter CJ, Kimoto E, Piotrowski M, Varma MV, et al. Quantitative assessment of the contribution of sodium-dependent taurocholate co-transporting polypeptide (NTCP) to the hepatic uptake of rosuvastatin, pitavastatin and fluvastatin. Biopharm Drug Dispos. 2013;34(8):452–61.PubMed
109.
Zurück zum Zitat Amundsen R, Christensen H, Zabihyan B, Åsberg A. Cyclosporine A, but not tacrolimus, shows relevant inhibition of organic anion-transporting protein 1B1-mediated transport of atorvastatin. Drug Metabol Dispos. 2010;38(9):1499–504. Amundsen R, Christensen H, Zabihyan B, Åsberg A. Cyclosporine A, but not tacrolimus, shows relevant inhibition of organic anion-transporting protein 1B1-mediated transport of atorvastatin. Drug Metabol Dispos. 2010;38(9):1499–504.
110.
Zurück zum Zitat Kato M, Shitara Y, Sato H, Yoshisue K, Hirano M, Ikeda T, et al. The quantitative prediction of CYP-mediated drug interaction by physiologically based pharmacokinetic modeling. Pharm Res. 2008;25(8):1891–901.PubMed Kato M, Shitara Y, Sato H, Yoshisue K, Hirano M, Ikeda T, et al. The quantitative prediction of CYP-mediated drug interaction by physiologically based pharmacokinetic modeling. Pharm Res. 2008;25(8):1891–901.PubMed
111.
Zurück zum Zitat Izumi S, Nozaki Y, Komori T, Maeda K, Takenaka O, Kusano K, et al. Substrate-dependent inhibition of organic anion transporting polypeptide 1B1: comparative analysis with prototypical probe substrates estradiol-17beta-glucuronide, estrone-3-sulfate, and sulfobromophthalein. Drug Metab Dispos. 2013;41(10):1859–66.PubMed Izumi S, Nozaki Y, Komori T, Maeda K, Takenaka O, Kusano K, et al. Substrate-dependent inhibition of organic anion transporting polypeptide 1B1: comparative analysis with prototypical probe substrates estradiol-17beta-glucuronide, estrone-3-sulfate, and sulfobromophthalein. Drug Metab Dispos. 2013;41(10):1859–66.PubMed
112.
Zurück zum Zitat Noe J, Portmann R, Brun ME, Funk C. Substrate-dependent drug–drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Drug Metab Dispos. 2007;35(8):1308–14.PubMed Noe J, Portmann R, Brun ME, Funk C. Substrate-dependent drug–drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Drug Metab Dispos. 2007;35(8):1308–14.PubMed
113.
Zurück zum Zitat Chu X, Korzekwa K, Elsby R, Fenner K, Galetin A, Lai Y, et al. Intracellular drug concentrations and transporters: measurement, modeling, and implications for the liver. Clin Pharmacol Ther. 2013;94(1):126–41.PubMedCentralPubMed Chu X, Korzekwa K, Elsby R, Fenner K, Galetin A, Lai Y, et al. Intracellular drug concentrations and transporters: measurement, modeling, and implications for the liver. Clin Pharmacol Ther. 2013;94(1):126–41.PubMedCentralPubMed
114.
Zurück zum Zitat Obach RS, Walsky RL, Venkatakrishnan K, Houston JB, Tremaine LM. In vitro cytochrome P450 inhibition data and the prediction of drug–drug interactions: qualitative relationships, quantitative predictions, and the rank-order approach. Clin Pharmacol Ther. 2005;78(6):582–92.PubMed Obach RS, Walsky RL, Venkatakrishnan K, Houston JB, Tremaine LM. In vitro cytochrome P450 inhibition data and the prediction of drug–drug interactions: qualitative relationships, quantitative predictions, and the rank-order approach. Clin Pharmacol Ther. 2005;78(6):582–92.PubMed
115.
Zurück zum Zitat Tamai I. Oral drug delivery utilizing intestinal OATP transporters. Adv Drug Deliv Rev. 2012;64(6):508–14.PubMed Tamai I. Oral drug delivery utilizing intestinal OATP transporters. Adv Drug Deliv Rev. 2012;64(6):508–14.PubMed
116.
Zurück zum Zitat 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(9):730–42.PubMed 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(9):730–42.PubMed
117.
Zurück zum Zitat Neuhoff S, Yeo KR, Barter Z, Jamei M, Turner DB, Rostami-Hodjegan A. Application of permeability-limited physiologically-based pharmacokinetic models: part I–digoxin pharmacokinetics incorporating P-glycoprotein-mediated efflux. J Pharm Sci. 2013;102(9):3145–60.PubMed Neuhoff S, Yeo KR, Barter Z, Jamei M, Turner DB, Rostami-Hodjegan A. Application of permeability-limited physiologically-based pharmacokinetic models: part I–digoxin pharmacokinetics incorporating P-glycoprotein-mediated efflux. J Pharm Sci. 2013;102(9):3145–60.PubMed
118.
Zurück zum Zitat Tachibana T, Kato M, Watanabe T, Mitsui T, Sugiyama Y. Method for predicting the risk of drug–drug interactions involving inhibition of intestinal CYP3A4 and P-glycoprotein. Xenobiotica. 2009;39(6):430–43.PubMed Tachibana T, Kato M, Watanabe T, Mitsui T, Sugiyama Y. Method for predicting the risk of drug–drug interactions involving inhibition of intestinal CYP3A4 and P-glycoprotein. Xenobiotica. 2009;39(6):430–43.PubMed
119.
Zurück zum Zitat Chow EC, Pang KS. Why we need proper PBPK models to examine intestine and liver oral drug absorption. Curr Drug Metab. 2013;14(1):57–79.PubMed Chow EC, Pang KS. Why we need proper PBPK models to examine intestine and liver oral drug absorption. Curr Drug Metab. 2013;14(1):57–79.PubMed
120.
Zurück zum Zitat Watanabe T, Kusuhara H, Watanabe T, Debori Y, Maeda K, Kondo T, et al. Prediction of the overall renal tubular secretion and hepatic clearance of anionic drugs and a renal drug–drug interaction involving organic anion transporter 3 in humans by in vitro uptake experiments. Drug Metab Dispos. 2011;39(6):1031–8.PubMed Watanabe T, Kusuhara H, Watanabe T, Debori Y, Maeda K, Kondo T, et al. Prediction of the overall renal tubular secretion and hepatic clearance of anionic drugs and a renal drug–drug interaction involving organic anion transporter 3 in humans by in vitro uptake experiments. Drug Metab Dispos. 2011;39(6):1031–8.PubMed
121.
Zurück zum Zitat Nakagomi-Hagihara R, Nakai D, Tokui T. Inhibition of human organic anion transporter 3 mediated pravastatin transport by gemfibrozil and the metabolites in humans. Xenobiotica. 2007;37(4):416–26.PubMed Nakagomi-Hagihara R, Nakai D, Tokui T. Inhibition of human organic anion transporter 3 mediated pravastatin transport by gemfibrozil and the metabolites in humans. Xenobiotica. 2007;37(4):416–26.PubMed
122.
Zurück zum Zitat Kyrklund C, Backman JT, Neuvonen M, Neuvonen PJ. Gemfibrozil increases plasma pravastatin concentrations and reduces pravastatin renal clearance. Clin Pharmacol Ther. 2003;73(6):538–44.PubMed Kyrklund C, Backman JT, Neuvonen M, Neuvonen PJ. Gemfibrozil increases plasma pravastatin concentrations and reduces pravastatin renal clearance. Clin Pharmacol Ther. 2003;73(6):538–44.PubMed
123.
Zurück zum Zitat Hsu V, de LTV M, Zhao P, Zhang L, Zheng JH, Nordmark A. Towards quantitation of the effects of renal impairment and probenecid inhibition on kidney uptake and efflux transporters, using physiologically based pharmacokinetic modelling and simulations. Clin Pharmacokinet. 2014;53(3):283–93.PubMedCentralPubMed Hsu V, de LTV M, Zhao P, Zhang L, Zheng JH, Nordmark A. Towards quantitation of the effects of renal impairment and probenecid inhibition on kidney uptake and efflux transporters, using physiologically based pharmacokinetic modelling and simulations. Clin Pharmacokinet. 2014;53(3):283–93.PubMedCentralPubMed
124.
Zurück zum Zitat 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(3):305–14.PubMedCentralPubMed 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(3):305–14.PubMedCentralPubMed
125.
Zurück zum Zitat 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(4):249–56.PubMed 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(4):249–56.PubMed
126.
Zurück zum Zitat 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(7):1279–89.PubMed 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(7):1279–89.PubMed
127.
Zurück zum Zitat Kajosaari LI, Niemi M, Neuvonen M, Laitila J, Neuvonen PJ, Backman JT. Cyclosporine markedly raises the plasma concentrations of repaglinide. Clin Pharmacol Ther. 2005;78(4):388–99.PubMed Kajosaari LI, Niemi M, Neuvonen M, Laitila J, Neuvonen PJ, Backman JT. Cyclosporine markedly raises the plasma concentrations of repaglinide. Clin Pharmacol Ther. 2005;78(4):388–99.PubMed
128.
Zurück zum Zitat Niemi M, Backman JT, Kajosaari LI, Leathart JB, Neuvonen M, Daly AK, et al. Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics. Clin Pharmacol Ther. 2005;77(6):468–78.PubMed Niemi M, Backman JT, Kajosaari LI, Leathart JB, Neuvonen M, Daly AK, et al. Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics. Clin Pharmacol Ther. 2005;77(6):468–78.PubMed
129.
Zurück zum Zitat Niemi M, Leathart JB, Neuvonen M, Backman JT, Daly AK, Neuvonen PJ. Polymorphism in CYP2C8 is associated with reduced plasma concentrations of repaglinide. Clin Pharmacol Ther. 2003;74(4):380–7.PubMed Niemi M, Leathart JB, Neuvonen M, Backman JT, Daly AK, Neuvonen PJ. Polymorphism in CYP2C8 is associated with reduced plasma concentrations of repaglinide. Clin Pharmacol Ther. 2003;74(4):380–7.PubMed
130.
Zurück zum Zitat Ogilvie BW, Zhang D, Li W, Rodrigues AD, Gipson AE, Holsapple J, et al. Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: implications for drug–drug interactions. Drug Metab Dispos. 2006;34(1):191–7.PubMed Ogilvie BW, Zhang D, Li W, Rodrigues AD, Gipson AE, Holsapple J, et al. Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: implications for drug–drug interactions. Drug Metab Dispos. 2006;34(1):191–7.PubMed
131.
Zurück zum Zitat 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(1):228–36.PubMed 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(1):228–36.PubMed
132.
Zurück zum Zitat Fujino H, Shimada S, Yamada I, Hirano M, Tsunenari Y, Kojima J. Studies on the interaction between fibrates and statins using human hepatic microsomes. Arzneimittelforschung. 2003;53(10):701–7.PubMed Fujino H, Shimada S, Yamada I, Hirano M, Tsunenari Y, Kojima J. Studies on the interaction between fibrates and statins using human hepatic microsomes. Arzneimittelforschung. 2003;53(10):701–7.PubMed
133.
Zurück zum Zitat 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(3):347–51.PubMed 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(3):347–51.PubMed
134.
Zurück zum Zitat Zheng HX, Huang Y, Frassetto LA, Benet LZ. 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. 2009;85(1):78–85.PubMedCentralPubMed Zheng HX, Huang Y, Frassetto LA, Benet LZ. 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. 2009;85(1):78–85.PubMedCentralPubMed
135.
Zurück zum Zitat Backman JT, Luurila H, Neuvonen M, Neuvonen PJ. Rifampin markedly decreases and gemfibrozil increases the plasma concentrations of atorvastatin and its metabolites. Clin Pharmacol Ther. 2005;78(2):154–67.PubMed Backman JT, Luurila H, Neuvonen M, Neuvonen PJ. Rifampin markedly decreases and gemfibrozil increases the plasma concentrations of atorvastatin and its metabolites. Clin Pharmacol Ther. 2005;78(2):154–67.PubMed
136.
Zurück zum Zitat Weber C, Schmitt R, Birnboeck H, Hopfgartner G, van Marle SP, Peeters PA, et al. Pharmacokinetics and pharmacodynamics of the endothelin-receptor antagonist bosentan in healthy human subjects. Clin Pharmacol Ther. 1996;60(2):124–37.PubMed Weber C, Schmitt R, Birnboeck H, Hopfgartner G, van Marle SP, Peeters PA, et al. Pharmacokinetics and pharmacodynamics of the endothelin-receptor antagonist bosentan in healthy human subjects. Clin Pharmacol Ther. 1996;60(2):124–37.PubMed
137.
Zurück zum Zitat Qiu X, Bi Y-A, Balogh LM, Lai Y. Absolute measurement of species differences in sodium taurocholate cotransporting polypeptide (NTCP/Ntcp) and its modulation in cultured hepatocytes. J Pharm Sci. 2013;102(9):3252–63.PubMed Qiu X, Bi Y-A, Balogh LM, Lai Y. Absolute measurement of species differences in sodium taurocholate cotransporting polypeptide (NTCP/Ntcp) and its modulation in cultured hepatocytes. J Pharm Sci. 2013;102(9):3252–63.PubMed
138.
Zurück zum Zitat Ho RH, Tirona RG, Leake BF, Glaeser H, Lee W, Lemke CJ, et al. Drug and bile acid transporters in rosuvastatin hepatic uptake: function, expression, and pharmacogenetics. Gastroenterology. 2006;130(6):1793–806.PubMed Ho RH, Tirona RG, Leake BF, Glaeser H, Lee W, Lemke CJ, et al. Drug and bile acid transporters in rosuvastatin hepatic uptake: function, expression, and pharmacogenetics. Gastroenterology. 2006;130(6):1793–806.PubMed
Metadaten
Titel
Prediction of Pharmacokinetics and Drug–Drug Interactions When Hepatic Transporters are Involved
verfasst von
Rui Li
Hugh A. Barton
Manthena V. Varma
Publikationsdatum
01.08.2014
Verlag
Springer International Publishing
Erschienen in
Clinical Pharmacokinetics / Ausgabe 8/2014
Print ISSN: 0312-5963
Elektronische ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-014-0156-z

Weitere Artikel der Ausgabe 8/2014

Clinical Pharmacokinetics 8/2014 Zur Ausgabe