Skip to main content
Top
Published in: Clinical Pharmacokinetics 6/2020

01-06-2020 | Breast Cancer | Original Research Article

Quantitative Prediction of Interactions Mediated by Transporters and Cytochromes: Application to Organic Anion Transporting Polypeptides, Breast Cancer Resistance Protein and Cytochrome 2C8

Authors: Michel Tod, Laurent Bourguignon, Nathalie Bleyzac, Sylvain Goutelle

Published in: Clinical Pharmacokinetics | Issue 6/2020

Login to get access

Abstract

Background

The in vivo mechanistic static model (IMSM) is an effective method to predict the magnitude of drug–drug interactions (DDIs) mediated by cytochromes.

Objective

The aim of this study was to extend the IMSM paradigm to DDIs mediated by organic anion transporting polypeptide (OATP) 1Bs, breast cancer resistance protein (BCRP) and cytochrome 2C8.

Methods

First, a generic model for this kind of interaction was established, and a literature search was then conducted to retrieve the area under the concentration–time curve (AUC) ratio of a large set of DDIs involving OATP1B1, OATP1B3, BCRP and cytochromes 2C8 or 3A4. The model was fitted to the data to estimate the characteristic parameters (contribution ratios [CRs] and inhibition or induction potencies [IXs]) by nonlinear regression, and the model was qualified by external validation on a different dataset. Lastly, the model was used to identify the risks of overexposure by DDIs of this type.

Results

A total of 27 substrates, 26 inhibitors, 3 inducers and 3 genetic variants were considered in the regression analysis. The number of observations (AUC ratios, denoted as Robs) was 101. Forty-six CRs and 47 IXs were estimated. The proportions of predictions within 0.67- to 1.5-fold and 0.5- to twofold Robs were 90% and 99%, respectively, for the internal validation, and 78% and 96%, respectively, for the external validation. The median fold-error was 1.03 (the ideal value is 1). The interquartile range of fold-error was 0.31, and the relative standard error of parameter estimates was, at most, 17%.

Conclusions

The IMSM approach was successfully extended to DDIs mediated by OATP1Bs, BCRP and cytochromes 2C8 or 3A4. The method revealed good predictive performances by internal and external validation.
Appendix
Available only for authorised users
Literature
1.
go back to reference International Transporter Consortium, Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36. International Transporter Consortium, Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36.
2.
go back to reference 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
3.
go back to reference 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
5.
go back to reference Wessler JD, Grip LT, Mendell J, Giugliano RP. The P-glycoprotein transport system and cardiovascular drugs. J Am Coll Cardiol. 2013;61(25):2495–502.PubMed Wessler JD, Grip LT, Mendell J, Giugliano RP. The P-glycoprotein transport system and cardiovascular drugs. J Am Coll Cardiol. 2013;61(25):2495–502.PubMed
6.
go back to reference Lee CA, O’Connor MA, Ritchie TK, Galetin A, Cook JA, Ragueneau-Majlessi I, et al. Breast cancer resistance protein (ABCG2) in clinical pharmacokinetics and drug interactions: practical recommendations for clinical victim and perpetrator drug-drug interaction study design. Drug Metab Dispos. 2015;43(4):490–509. Lee CA, O’Connor MA, Ritchie TK, Galetin A, Cook JA, Ragueneau-Majlessi I, et al. Breast cancer resistance protein (ABCG2) in clinical pharmacokinetics and drug interactions: practical recommendations for clinical victim and perpetrator drug-drug interaction study design. Drug Metab Dispos. 2015;43(4):490–509.
7.
go back to reference Mao Q, Unadkat JD. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport–an update. AAPS J. 2015;17(1):65–82.PubMed Mao Q, Unadkat JD. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport–an update. AAPS J. 2015;17(1):65–82.PubMed
8.
go back to reference Us FDA. Guidance for industry: drug interaction studies—study design, data analysis, implications for dosing, and labeling recommendations. Rockville: US FDA; 2012. Us FDA. Guidance for industry: drug interaction studies—study design, data analysis, implications for dosing, and labeling recommendations. Rockville: US FDA; 2012.
10.
go back to reference 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
11.
go back to reference 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
12.
go back to reference Elsby R, Hilgendorf C, Fenner K. Understanding the critical disposition pathways of statins to assess drug-drug interaction risk during drug development: it’s not just about OATP1B1. Clin Pharmacol Ther. 2012;92(5):584–98.PubMed Elsby R, Hilgendorf C, Fenner K. Understanding the critical disposition pathways of statins to assess drug-drug interaction risk during drug development: it’s not just about OATP1B1. Clin Pharmacol Ther. 2012;92(5):584–98.PubMed
13.
go back to reference Pfeifer ND, Goss SL, Swift B, Ghibellini G, Ivanovic M, Heizer WD, et al. Effect of ritonavir on (99m)technetium-mebrofenin disposition in humans: a semi-PBPK modeling and in vitro approach to predict transporter-mediated DDIs. CPT Pharmacometr Syst Pharmacol. 2013;2:e20. Pfeifer ND, Goss SL, Swift B, Ghibellini G, Ivanovic M, Heizer WD, et al. Effect of ritonavir on (99m)technetium-mebrofenin disposition in humans: a semi-PBPK modeling and in vitro approach to predict transporter-mediated DDIs. CPT Pharmacometr Syst Pharmacol. 2013;2:e20.
14.
go back to reference 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
15.
go back to reference Li R, Barton HA, Varma MV. Prediction of pharmacokinetics and drug-drug interactions when hepatic transporters are involved. Clin Pharmacokinet. 2014;53(8):659–78.PubMed Li R, Barton HA, Varma MV. Prediction of pharmacokinetics and drug-drug interactions when hepatic transporters are involved. Clin Pharmacokinet. 2014;53(8):659–78.PubMed
16.
go back to reference Maeda K. Organic anion transporting polypeptide (OATP)1B1 and OATP1B3 as important regulators of the pharmacokinetics of substrate drugs. Biol Pharm Bull. 2015;38(2):155–68.PubMed Maeda K. Organic anion transporting polypeptide (OATP)1B1 and OATP1B3 as important regulators of the pharmacokinetics of substrate drugs. Biol Pharm Bull. 2015;38(2):155–68.PubMed
17.
go back to reference Yoshikado T, Maeda K, Kusuhara H, Furihata KI, Sugiyama Y. Quantitative analyses of the influence of parameters governing rate-determining process of hepatic elimination of drugs on the magnitudes of drug–drug interactions via hepatic OATPs and CYP3A using physiologically based pharmacokinetic models. J Pharm Sci. 2017;106(9):2739–50.PubMed Yoshikado T, Maeda K, Kusuhara H, Furihata KI, Sugiyama Y. Quantitative analyses of the influence of parameters governing rate-determining process of hepatic elimination of drugs on the magnitudes of drug–drug interactions via hepatic OATPs and CYP3A using physiologically based pharmacokinetic models. J Pharm Sci. 2017;106(9):2739–50.PubMed
18.
go back to reference Naritomi Y, Terashita S, Kimura S, Suzuki A, Kagayama A, Sugiyama Y. Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans. Drug Metab Dispos. 2001;29(10):1316–24.PubMed Naritomi Y, Terashita S, Kimura S, Suzuki A, Kagayama A, Sugiyama Y. Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans. Drug Metab Dispos. 2001;29(10):1316–24.PubMed
19.
go back to reference Durmus S, van Hoppe S, Schinkel AH. The impact of Organic Anion-Transporting Polypeptides (OATPs) on disposition and toxicity of antitumor drugs: insights from knockout and humanized mice. Drug Resist Updat. 2016;27:72–88.PubMed Durmus S, van Hoppe S, Schinkel AH. The impact of Organic Anion-Transporting Polypeptides (OATPs) on disposition and toxicity of antitumor drugs: insights from knockout and humanized mice. Drug Resist Updat. 2016;27:72–88.PubMed
20.
go back to reference Vaidyanathan J, Yoshida K, Arya V, Zhang L. Comparing various in vitro prediction criteria to assess the potential of a new molecular entity to inhibit organic anion transporting polypeptide 1B1. J Clin Pharmacol. 2016;56(Suppl 7):S59–72.PubMed Vaidyanathan J, Yoshida K, Arya V, Zhang L. Comparing various in vitro prediction criteria to assess the potential of a new molecular entity to inhibit organic anion transporting polypeptide 1B1. J Clin Pharmacol. 2016;56(Suppl 7):S59–72.PubMed
21.
go back to reference Miyauchi S, Masuda M, Kim SJ, Tanaka Y, Lee KR, Iwakado S, et al. The phenomenon of albumin-mediated hepatic uptake of organic anion transport polypeptide substrates: prediction of the in vivo uptake clearance from the in vitro uptake by isolated hepatocytes using a facilitated-dissociation model. Drug Metab Dispos. 2018;46(3):259–67.PubMed Miyauchi S, Masuda M, Kim SJ, Tanaka Y, Lee KR, Iwakado S, et al. The phenomenon of albumin-mediated hepatic uptake of organic anion transport polypeptide substrates: prediction of the in vivo uptake clearance from the in vitro uptake by isolated hepatocytes using a facilitated-dissociation model. Drug Metab Dispos. 2018;46(3):259–67.PubMed
22.
go back to reference Bowman CM, Benet LZ. In vitro-in vivo extrapolation and hepatic clearance-dependent underprediction. J Pharm Sci. 2019;108(7):2500–4.PubMed Bowman CM, Benet LZ. In vitro-in vivo extrapolation and hepatic clearance-dependent underprediction. J Pharm Sci. 2019;108(7):2500–4.PubMed
23.
go back to reference Wood FL, Houston JB, Hallifax D. Clearance prediction methodology needs fundamental improvement: trends common to rat and human hepatocytes/microsomes and implications for experimental methodology. Drug Metab Dispos. 2017;45(11):1178–88.PubMed Wood FL, Houston JB, Hallifax D. Clearance prediction methodology needs fundamental improvement: trends common to rat and human hepatocytes/microsomes and implications for experimental methodology. Drug Metab Dispos. 2017;45(11):1178–88.PubMed
24.
go back to reference Ohno Y, Hisaka A, Suzuki H. General framework for the quantitative prediction of CYP3A4-mediated oral drug interactions based on the AUC increase by coadministration of standard drugs. Clin Pharmacokinet. 2007;46(8):681–96.PubMed Ohno Y, Hisaka A, Suzuki H. General framework for the quantitative prediction of CYP3A4-mediated oral drug interactions based on the AUC increase by coadministration of standard drugs. Clin Pharmacokinet. 2007;46(8):681–96.PubMed
25.
go back to reference Ohno Y, Hisaka A, Ueno M, Suzuki H. General framework for the prediction of oral drug interactions caused by CYP3A4 induction from in vivo information. Clin Pharmacokinet. 2008;47(10):669–80.PubMed Ohno Y, Hisaka A, Ueno M, Suzuki H. General framework for the prediction of oral drug interactions caused by CYP3A4 induction from in vivo information. Clin Pharmacokinet. 2008;47(10):669–80.PubMed
26.
go back to reference Castellan AC, Tod M, Gueyffier F, Audars M, Cambriels F, Kassai B, et al. Quantitative prediction of the impact of drug interactions and genetic polymorphisms on cytochrome P450 2C9 substrate exposure. Clin Pharmacokinet. 2013;52(3):199–209.PubMed Castellan AC, Tod M, Gueyffier F, Audars M, Cambriels F, Kassai B, et al. Quantitative prediction of the impact of drug interactions and genetic polymorphisms on cytochrome P450 2C9 substrate exposure. Clin Pharmacokinet. 2013;52(3):199–209.PubMed
27.
go back to reference Gabriel L, Tod M, Goutelle S. Quantitative prediction of drug interactions caused by CYP1A2 inhibitors and inducers. Clin Pharmacokinet. 2016;55(8):977–90.PubMed Gabriel L, Tod M, Goutelle S. Quantitative prediction of drug interactions caused by CYP1A2 inhibitors and inducers. Clin Pharmacokinet. 2016;55(8):977–90.PubMed
28.
go back to reference Goutelle S, Bourguignon L, Bleyzac N, Berry J, Clavel-Grabit F, Tod M. In vivo quantitative prediction of the effect of gene polymorphisms and drug interactions on drug exposure for CYP2C19 substrates. AAPS J. 2013;15(2):415–26.PubMedPubMedCentral Goutelle S, Bourguignon L, Bleyzac N, Berry J, Clavel-Grabit F, Tod M. In vivo quantitative prediction of the effect of gene polymorphisms and drug interactions on drug exposure for CYP2C19 substrates. AAPS J. 2013;15(2):415–26.PubMedPubMedCentral
29.
go back to reference Tod M, Goutelle S, Clavel-Grabit F, Nicolas G, Charpiat B. Quantitative prediction of cytochrome P450 (CYP) 2D6-mediated drug interactions. Clin Pharmacokinet. 2011;50(8):519–30.PubMed Tod M, Goutelle S, Clavel-Grabit F, Nicolas G, Charpiat B. Quantitative prediction of cytochrome P450 (CYP) 2D6-mediated drug interactions. Clin Pharmacokinet. 2011;50(8):519–30.PubMed
30.
go back to reference Tod M, Nkoud-Mongo C, Gueyffier F. Impact of genetic polymorphism on drug-drug interactions mediated by cytochromes: a general approach. AAPS J. 2013;15(4):1242–52.PubMedPubMedCentral Tod M, Nkoud-Mongo C, Gueyffier F. Impact of genetic polymorphism on drug-drug interactions mediated by cytochromes: a general approach. AAPS J. 2013;15(4):1242–52.PubMedPubMedCentral
31.
go back to reference Tod M, Goutelle S, Bleyzac N, Bourguignon L. A generic model for quantitative prediction of interactions mediated by efflux transporters and cytochromes: application to P-glycoprotein and cytochrome 3A4. Clin Pharmacokinet. 2019;58(4):503–23.PubMed Tod M, Goutelle S, Bleyzac N, Bourguignon L. A generic model for quantitative prediction of interactions mediated by efflux transporters and cytochromes: application to P-glycoprotein and cytochrome 3A4. Clin Pharmacokinet. 2019;58(4):503–23.PubMed
32.
go back to reference Tod M, Pierrillas PB, Bourguignon L, Goutelle S. Comparison of the static in vivo approach to a physiologically based pharmacokinetic approach for metabolic drug–drug interactions prediction. International Journal of Pharmacokinetics. https://doi.org/10.4155/ipk.16.2(Epub 4 Apr 2016). Tod M, Pierrillas PB, Bourguignon L, Goutelle S. Comparison of the static in vivo approach to a physiologically based pharmacokinetic approach for metabolic drug–drug interactions prediction. International Journal of Pharmacokinetics. https://​doi.​org/​10.​4155/​ipk.​16.​2(Epub 4 Apr 2016).
33.
go back to reference 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
34.
go back to reference Loue C, Tod M. Reliability and extension of quantitative prediction of CYP3A4-mediated drug interactions based on clinical data. AAPS J. 2014;16(6):1309–20.PubMedPubMedCentral Loue C, Tod M. Reliability and extension of quantitative prediction of CYP3A4-mediated drug interactions based on clinical data. AAPS J. 2014;16(6):1309–20.PubMedPubMedCentral
35.
go back to reference Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of cytochrome P450 2C8 in drug metabolism and interactions. Pharmacol Rev. 2016;68(1):168–241.PubMed Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of cytochrome P450 2C8 in drug metabolism and interactions. Pharmacol Rev. 2016;68(1):168–241.PubMed
36.
go back to reference He YJ, Zhang W, Chen Y, Guo D, Tu JH, Xu LY, et al. Rifampicin alters atorvastatin plasma concentration on the basis of SLCO1B1 521T > C polymorphism. Clin Chim Acta. 2009;405(1–2):49–52.PubMed He YJ, Zhang W, Chen Y, Guo D, Tu JH, Xu LY, et al. Rifampicin alters atorvastatin plasma concentration on the basis of SLCO1B1 521T > C polymorphism. Clin Chim Acta. 2009;405(1–2):49–52.PubMed
37.
go back to reference Tod M, Bourguignon L, Bleyzac N, Goutelle S. A model for predicting the interindividual variability of drug–drug interactions. AAPS J. 2017;19(2):497–509.PubMed Tod M, Bourguignon L, Bleyzac N, Goutelle S. A model for predicting the interindividual variability of drug–drug interactions. AAPS J. 2017;19(2):497–509.PubMed
38.
go back to reference Tod M, Goutelle S, Gagnieu MC, Genophar IIWG. Genotype-based quantitative prediction of drug exposure for drugs metabolized by CYP2D6. Clin Pharmacol Ther. 2011;90(4):582–7.PubMed Tod M, Goutelle S, Gagnieu MC, Genophar IIWG. Genotype-based quantitative prediction of drug exposure for drugs metabolized by CYP2D6. Clin Pharmacol Ther. 2011;90(4):582–7.PubMed
39.
go back to reference Yoshikado T, Maeda K, Furihata S, Terashima H, Nakayama T, Ishigame K, et al. A clinical cassette dosing study for evaluating the contribution of hepatic OATPs and CYP3A to drug–drug interactions. Pharm Res. 2017;34(8):1570–83.PubMed Yoshikado T, Maeda K, Furihata S, Terashima H, Nakayama T, Ishigame K, et al. A clinical cassette dosing study for evaluating the contribution of hepatic OATPs and CYP3A to drug–drug interactions. Pharm Res. 2017;34(8):1570–83.PubMed
40.
go back to reference Yoshida K, Guo C, Sane R. Quantitative prediction of OATP-mediated drug–drug interactions with model-based analysis of endogenous biomarker kinetics. CPT Pharmacometrics Syst Pharmacol. 2018;7(8):517–24.PubMedPubMedCentral Yoshida K, Guo C, Sane R. Quantitative prediction of OATP-mediated drug–drug interactions with model-based analysis of endogenous biomarker kinetics. CPT Pharmacometrics Syst Pharmacol. 2018;7(8):517–24.PubMedPubMedCentral
41.
go back to reference Kunze A, Ediage EN, Dillen L, Monshouwer M, Snoeys J. Clinical investigation of coproporphyrins as sensitive biomarkers to predict mild to strong OATP1B-mediated drug–drug interactions. Clin Pharmacokinet. 2018;57(12):1559–70.PubMed Kunze A, Ediage EN, Dillen L, Monshouwer M, Snoeys J. Clinical investigation of coproporphyrins as sensitive biomarkers to predict mild to strong OATP1B-mediated drug–drug interactions. Clin Pharmacokinet. 2018;57(12):1559–70.PubMed
42.
go back to reference 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
43.
go back to reference 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
44.
go back to reference 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.PubMedPubMedCentral 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.PubMedPubMedCentral
45.
go back to reference Muck W, Mai I, Fritsche L, Ochmann K, Rohde G, Unger S, et al. Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporine-treated kidney transplant recipients. Clin Pharmacol Ther. 1999;65(3):251–61.PubMed Muck W, Mai I, Fritsche L, Ochmann K, Rohde G, Unger S, et al. Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporine-treated kidney transplant recipients. Clin Pharmacol Ther. 1999;65(3):251–61.PubMed
46.
go back to reference Itkonen MK, Tornio A, Neuvonen M, Neuvonen PJ, Niemi M, Backman JT. Clopidogrel and gemfibrozil strongly inhibit the CYP2C8-dependent formation of 3-hydroxydesloratadine and increase desloratadine exposure in humans. Drug Metab Dispos. 2019;47(4):377–85.PubMed Itkonen MK, Tornio A, Neuvonen M, Neuvonen PJ, Niemi M, Backman JT. Clopidogrel and gemfibrozil strongly inhibit the CYP2C8-dependent formation of 3-hydroxydesloratadine and increase desloratadine exposure in humans. Drug Metab Dispos. 2019;47(4):377–85.PubMed
47.
go back to reference Macha S, Koenen R, Sennewald R, Schone K, Hummel N, Riedmaier S, et al. Effect of gemfibrozil, rifampicin, or probenecid on the pharmacokinetics of the SGLT2 inhibitor empagliflozin in healthy volunteers. Clin Ther. 2014;36(2):280–90e1. Macha S, Koenen R, Sennewald R, Schone K, Hummel N, Riedmaier S, et al. Effect of gemfibrozil, rifampicin, or probenecid on the pharmacokinetics of the SGLT2 inhibitor empagliflozin in healthy volunteers. Clin Ther. 2014;36(2):280–90e1.
48.
go back to reference Varma MV, Kimoto E, Scialis R, Bi Y, Lin J, Eng H, et al. Transporter-mediated hepatic uptake plays an important role in the pharmacokinetics and drug–drug interactions of montelukast. Clin Pharmacol Ther. 2017;101(3):406–15.PubMed Varma MV, Kimoto E, Scialis R, Bi Y, Lin J, Eng H, et al. Transporter-mediated hepatic uptake plays an important role in the pharmacokinetics and drug–drug interactions of montelukast. Clin Pharmacol Ther. 2017;101(3):406–15.PubMed
49.
go back to reference Kim SJ, Yoshikado T, Ieiri I, Maeda K, Kimura M, Irie S, et al. Clarification of the mechanism of clopidogrel-mediated drug–drug interaction in a clinical cassette small-dose study and its prediction based on in vitro information. Drug Metab Dispos. 2016;44(10):1622–32.PubMed Kim SJ, Yoshikado T, Ieiri I, Maeda K, Kimura M, Irie S, et al. Clarification of the mechanism of clopidogrel-mediated drug–drug interaction in a clinical cassette small-dose study and its prediction based on in vitro information. Drug Metab Dispos. 2016;44(10):1622–32.PubMed
50.
go back to reference Niemi M, Neuvonen PJ, Kivisto KT. The cytochrome P4503A4 inhibitor clarithromycin increases the plasma concentrations and effects of repaglinide. Clin Pharmacol Ther. 2001;70(1):58–65.PubMed Niemi M, Neuvonen PJ, Kivisto KT. The cytochrome P4503A4 inhibitor clarithromycin increases the plasma concentrations and effects of repaglinide. Clin Pharmacol Ther. 2001;70(1):58–65.PubMed
51.
go back to reference 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
52.
go back to reference Tornio A, Filppula AM, Kailari O, Neuvonen M, Nyronen TH, Tapaninen T, 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(4):498–507.PubMed Tornio A, Filppula AM, Kailari O, Neuvonen M, Nyronen TH, Tapaninen T, 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(4):498–507.PubMed
53.
go back to reference Niemi M, Kajosaari LI, Neuvonen M, Backman JT, Neuvonen PJ. The CYP2C8 inhibitor trimethoprim increases the plasma concentrations of repaglinide in healthy subjects. Br J Clin Pharmacol. 2004;57(4):441–7.PubMedPubMedCentral Niemi M, Kajosaari LI, Neuvonen M, Backman JT, Neuvonen PJ. The CYP2C8 inhibitor trimethoprim increases the plasma concentrations of repaglinide in healthy subjects. Br J Clin Pharmacol. 2004;57(4):441–7.PubMedPubMedCentral
54.
go back to reference Niemi M, Backman JT, Neuvonen PJ. Effects of trimethoprim and rifampin on the pharmacokinetics of the cytochrome P450 2C8 substrate rosiglitazone. Clin Pharmacol Ther. 2004;76(3):239–49.PubMed Niemi M, Backman JT, Neuvonen PJ. Effects of trimethoprim and rifampin on the pharmacokinetics of the cytochrome P450 2C8 substrate rosiglitazone. Clin Pharmacol Ther. 2004;76(3):239–49.PubMed
55.
go back to reference Backman JT, Kyrklund C, Kivisto KT, Wang JS, Neuvonen PJ. Plasma concentrations of active simvastatin acid are increased by gemfibrozil. Clin Pharmacol Ther. 2000;68(2):122–9.PubMed Backman JT, Kyrklund C, Kivisto KT, Wang JS, Neuvonen PJ. Plasma concentrations of active simvastatin acid are increased by gemfibrozil. Clin Pharmacol Ther. 2000;68(2):122–9.PubMed
56.
go back to reference 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
57.
go back to reference Kawai R, Mathew D, Tanaka C, Rowland M. Physiologically based pharmacokinetics of cyclosporine A: extension to tissue distribution kinetics in rats and scale-up to human. J Pharmacol Exp Ther. 1998;287(2):457–68.PubMed Kawai R, Mathew D, Tanaka C, Rowland M. Physiologically based pharmacokinetics of cyclosporine A: extension to tissue distribution kinetics in rats and scale-up to human. J Pharmacol Exp Ther. 1998;287(2):457–68.PubMed
Metadata
Title
Quantitative Prediction of Interactions Mediated by Transporters and Cytochromes: Application to Organic Anion Transporting Polypeptides, Breast Cancer Resistance Protein and Cytochrome 2C8
Authors
Michel Tod
Laurent Bourguignon
Nathalie Bleyzac
Sylvain Goutelle
Publication date
01-06-2020
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 6/2020
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-019-00853-2

Other articles of this Issue 6/2020

Clinical Pharmacokinetics 6/2020 Go to the issue