Effect of P-glycoprotein (P-gp) Inducers on Exposure of P-gp Substrates: Review of Clinical Drug-Drug Interaction Studies

Mohamed Elmeliegy, Manoli Vourvahis, Cen Guo, Diane D Wang, Mohamed Elmeliegy, Manoli Vourvahis, Cen Guo, Diane D Wang

Abstract

Understanding transporter-mediated drug-drug interactions (DDIs) for investigational agents is important during drug development to assess DDI liability, its clinical relevance, and to determine appropriate DDI management strategies. P-glycoprotein (P-gp) is an efflux transporter that influences the pharmacokinetics (PK) of various compounds. Assessing transporter induction in vitro is challenging and is not always predictive of in vivo effects, and hence there is a need to consider clinical DDI studies; however, there is no clear guidance on when clinical evaluation of transporter induction is required. Furthermore, there is no proposed list of index transporter inducers to be used in clinical studies. This review evaluated DDI studies with known P-gp inducers to better understand the mechanism and site of P-gp induction, as well as the magnitude of induction effect on the exposure of P-gp substrates. Our review indicates that P-gp and cytochrome P450 (CYP450) enzymes are co-regulated via the pregnane xenobiotic receptor (PXR) and the constitutive androstane receptor (CAR). The magnitude of the decrease in substrate drug exposure by P-gp induction is generally less than that of CYP3A. Most P-gp inducers reduced total bioavailability with a minor impact on renal clearance, despite known expression of P-gp at the apical membrane of the kidney proximal tubules. Rifampin is the most potent P-gp inducer, resulting in an average reduction in substrate exposure ranging between 20 and 67%. For other inducers, the reduction in P-gp substrate exposure ranged from 12 to 42%. A lower reduction in exposure of the P-gp substrate was observed with a lower dose of the inducer and/or if the administration of the inducer and substrate was simultaneous, i.e. not staggered. These findings suggest that clinical evaluation of the impact of P-gp inducers on the PK of investigational agents that are substrates for P-gp might be warranted only for compounds with a relatively steep exposure-efficacy relationship.

Conflict of interest statement

Mohamed Elmeliegy, Manoli Vourvahis, Cen Guo, and Diane D. Wang are employees of Pfizer and receive stock and stock options as part of their employment.

References

    1. Tanigawara Y. Role of P-glycoprotein in drug disposition. Ther Drug Monit. 2000;22(1):137–140.
    1. Fromm MF. P-glycoprotein: a defense mechanism limiting oral bioavailability and CNS accumulation of drugs. Int J Clin Pharmacol Ther. 2000;38(2):69–74.
    1. Fromm MF. Importance of P-glycoprotein at blood–tissue barriers. Trends Pharmacol Sci. 2004;25(8):423–429.
    1. Lin JH, Yamazaki M. Role of P-glycoprotein in pharmacokinetics: clinical implications. Clin Pharmacokinet. 2003;42(1):59–98.
    1. Lund M, Petersen TS, Dalhoff KP. Clinical implications of P-glycoprotein modulation in drug–drug interactions. Drugs. 2017;77(8):859–883.
    1. FDA, Clinical Drug Interaction Studies—Study Design, Data Analysis, and Clinical Implications—Guidance for Industry; 2017.
    1. Ahlin G, et al. Endogenous gene and protein expression of drug-transporting proteins in cell lines routinely used in drug discovery programs. Drug Metab Dispos. 2009;37(12):2275–2283.
    1. Thompson CG, et al. Multimodal analysis of drug transporter expression in gastrointestinal tissue. AIDS. 2017;31(12):1669–1678.
    1. Ohtsuki S, et al. Simultaneous absolute protein quantification of transporters, cytochromes P450, and UDP-glucuronosyltransferases as a novel approach for the characterization of individual human liver: comparison with mRNA levels and activities. Drug Metab Dispos. 2012;40(1):83–92.
    1. Brueck S, et al. Transcriptional and post-transcriptional regulation of duodenal P-glycoprotein and MRP2 in healthy human subjects after chronic treatment with rifampin and carbamazepine. Mol Pharm. 2019;16(9):3823–3830.
    1. Bruckmueller H, et al. Clinically relevant multidrug transporters are regulated by microRNAs along the human intestine. Mol Pharm. 2017;14(7):2245–2253.
    1. Guo C, et al. Farnesoid X receptor agonists obeticholic acid and chenodeoxycholic acid increase bile acid efflux in sandwich-cultured human hepatocytes: functional evidence and mechanisms. J Pharmacol Exp Ther. 2018;365(2):413–421.
    1. Gu X, Manautou JE. Regulation of hepatic ABCC transporters by xenobiotics and in disease states. Drug Metab Rev. 2010;42(3):482–538.
    1. Schonhoff CM, Webster CR, Anwer MS. Taurolithocholate-induced MRP2 retrieval involves MARCKS phosphorylation by protein kinase C in HUH-NTCP Cells. Hepatology. 2013;58(1):284–292.
    1. Chandra P, et al. Modulation of hepatic canalicular or basolateral transport proteins alters hepatobiliary disposition of a model organic anion in the isolated perfused rat liver. Drug Metab Dispos. 2005;33(8):1238–1243.
    1. Zhang P, et al. Role of glycosylation in trafficking of Mrp2 in sandwich-cultured rat hepatocytes. Mol Pharmacol. 2005;67(4):1334–1341.
    1. Kirby BJ, et al. Complex drug interactions of the HIV protease inhibitors 3: effect of simultaneous or staggered dosing of digoxin and ritonavir, nelfinavir, rifampin, or bupropion. Drug Metab Dispos. 2012;40(3):610–616.
    1. Reitman ML, et al. Rifampin’s acute inhibitory and chronic inductive drug interactions: experimental and model-based approaches to drug–drug interaction trial design. Clin Pharmacol Ther. 2011;89(2):234–242.
    1. Collins JA, Fauser BC. Balancing the strengths of systematic and narrative reviews. Hum Reprod Update. 2005;11(2):103–104.
    1. Wang YM, et al. Role of CAR and PXR in xenobiotic sensing and metabolism. Expert Opin Drug Metab Toxicol. 2012;8(7):803–817.
    1. Kliewer SA, et al. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell. 1998;92(1):73–82.
    1. Goodwin B, Hodgson E, Liddle C. The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Mol Pharmacol. 1999;56(6):1329–1339.
    1. Geick A, Eichelbaum M, Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem. 2001;276(18):14581–14587.
    1. Olinga P, et al. Coordinated induction of drug transporters and phase I and II metabolism in human liver slices. Eur J Pharm Sci. 2008;33(4–5):380–389.
    1. Lehmann JM, et al. The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Investig. 1998;102(5):1016–1023.
    1. Ferguson SS, et al. Regulation of human CYP2C9 by the constitutive androstane receptor: discovery of a new distal binding site. Mol Pharmacol. 2002;62(3):737–746.
    1. Chen Y, et al. Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. J Pharmacol Exp Ther. 2004;308(2):495–501.
    1. Liu H, Sahi J. Role of hepatic drug transporters in drug development. J Clin Pharmacol. 2016;56(Suppl 7):S11–S22.
    1. Patel M, Taskar KS, Zamek-Gliszczynski MJ. Importance of hepatic transporters in clinical disposition of drugs and their metabolites. J Clin Pharmacol. 2016;56(Suppl 7):S23–S39.
    1. Meyer zu Schwabedissen HE, et al. Interplay between the nuclear receptor pregnane X receptor and the uptake transporter organic anion transporter polypeptide 1A2 selectively enhances estrogen effects in breast cancer. Cancer Res. 2008;68(22):9338–9347.
    1. Benson EA, et al. Rifampin regulation of drug transporters gene expression and the association of MicroRNAs in human hepatocytes. Front Pharmacol. 2016;7:111.
    1. FDA, Drug development and drug interactions: table of substrates, inhibitors and inducers; 2017.
    1. Shugarts S, Benet LZ. The role of transporters in the pharmacokinetics of orally administered drugs. Pharm Res. 2009;26(9):2039–2054.
    1. Benet LZ, Broccatelli F, Oprea TI. BDDCS applied to over 900 drugs. AAPS J. 2011;13(4):519–547.
    1. Hosey CM, Chan R, Benet LZ. BDDCS predictions, self-correcting aspects of BDDCS assignments, BDDCS assignment corrections, and classification for more than 175 additional drugs. AAPS J. 2016;18(1):251–260.
    1. Kullak-Ublick GA, et al. Organic anion-transporting polypeptide B (OATP-B) and its functional comparison with three other OATPs of human liver. Gastroenterology. 2001;120(2):525–533.
    1. Taub ME, et al. Digoxin is not a substrate for organic anion-transporting polypeptide transporters OATP1A2, OATP1B1, OATP1B3, and OATP2B1 but is a substrate for a sodium-dependent transporter expressed in HEK293 cells. Drug Metab Dispos. 2011;39(11):2093–2102.
    1. Seward DJ, et al. Functional complementation between a novel mammalian polygenic transport complex and an evolutionarily ancient organic solute transporter, OSTalpha-OSTbeta. J Biol Chem. 2003;278(30):27473–27482.
    1. Malinen MM, et al. Organic solute transporter OSTalpha/beta is overexpressed in nonalcoholic steatohepatitis and modulated by drugs associated with liver injury. Am J Physiol Gastrointest Liver Physiol. 2018;314(5):G597–g609.
    1. Tahara H, et al. P-glycoprotein plays a major role in the efflux of fexofenadine in the small intestine and blood–brain barrier, but only a limited role in its biliary excretion. Drug Metab Dispos. 2005;33(7):963–968.
    1. Swift B, Tian X, Brouwer KL. Integration of preclinical and clinical data with pharmacokinetic modeling and simulation to evaluate fexofenadine as a probe for hepatobiliary transport function. Pharm Res. 2009;26(8):1942–1951.
    1. Tian X, et al. Multidrug resistance-associated protein 2 is primarily responsible for the biliary excretion of fexofenadine in mice. Drug Metab Dispos. 2008;36(1):61–64.
    1. Matsushima S, et al. The inhibition of human multidrug and toxin extrusion 1 is involved in the drug–drug interaction caused by cimetidine. Drug Metab Dispos. 2009;37(3):555–559.
    1. Matsushima S, et al. Involvement of multiple efflux transporters in hepatic disposition of fexofenadine. Mol Pharmacol. 2008;73(5):1474–1483.
    1. Cvetkovic M, et al. OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine. Drug Metab Dispos. 1999;27(8):866–871.
    1. Nozawa T, et al. Functional characterization of pH-sensitive organic anion transporting polypeptide OATP-B in human. J Pharmacol Exp Ther. 2004;308(2):438–445.
    1. Shimizu M, et al. Contribution of OATP (organic anion-transporting polypeptide) family transporters to the hepatic uptake of fexofenadine in humans. Drug Metab Dispos. 2005;33(10):1477–1481.
    1. Tian X, et al. Impact of basolateral multidrug resistance-associated protein (Mrp) 3 and Mrp4 on the hepatobiliary disposition of fexofenadine in perfused mouse livers. Drug Metab Dispos. 2008;36(5):911–915.
    1. Moj D, et al. A comprehensive whole-body physiologically based pharmacokinetic model of dabigatran etexilate, dabigatran and dabigatran glucuronide in healthy adults and renally impaired patients. Clin Pharmacokinet. 2019;58:1577–1593.
    1. Tsuruya Y, et al. Different involvement of OAT in renal disposition of oral anticoagulants rivaroxaban, dabigatran, and apixaban. J Pharm Sci. 2017;106(9):2524–2534.
    1. Shen H, et al. Renal excretion of dabigatran: the potential role of multidrug and toxin extrusion (MATE) proteins. Mol Pharm. 2019;16(9):4065–4076.
    1. Stangier J, et al. Pharmacokinetics and pharmacodynamics of the direct oral thrombin inhibitor dabigatran in healthy elderly subjects. Clin Pharmacokinet. 2008;47(1):47–59.
    1. Troutman MD, Thakker DR. Efflux ratio cannot assess P-glycoprotein-mediated attenuation of absorptive transport: asymmetric effect of P-glycoprotein on absorptive and secretory transport across Caco-2 cell monolayers. Pharm Res. 2003;20(8):1200–1209.
    1. Shirasaka Y, et al. Species difference in the effect of grapefruit juice on intestinal absorption of talinolol between human and rat. J Pharmacol Exp Ther. 2010;332(1):181–189.
    1. Aronson JK. Clinical pharmacokinetics of digoxin 1980. Clin Pharmacokinet. 1980;5(2):137–149.
    1. US FDA. LANOXIN (Digoxin) Tablets. US FDA Product Insert.
    1. Molimard M, Diquet B, Benedetti MS. Comparison of pharmacokinetics and metabolism of desloratadine, fexofenadine, levocetirizine and mizolastine in humans. Fundam Clin Pharmacol. 2004;18(4):399–411.
    1. US FDA. ALLEGRA (fexofenadine hydrochloride) Capsules and Tablets. US FDA Product Insert.
    1. US FDA. PRADAXA® (dabigatran etexilate mesylate) capsules for oral use. US FDA Product Insert; 2010.
    1. Terhaag B, et al. The biliary elimination of the selective beta-receptor blocking drug talinolol in man. Int J Clin Pharmacol Ther Toxicol. 1989;27(4):170–172.
    1. Haustein KO, Fritzsche K. On the pharmacokinetics of talinolol, a new beta 1-receptor blocking agent. Int J Clin Pharmacol Ther Toxicol. 1981;19(9):392–395.
    1. Acocella G. Clinical pharmacokinetics of rifampicin. Clin Pharmacokinet. 1978;3(2):108–127.
    1. Westphal K, et al. Induction of P-glycoprotein by rifampin increases intestinal secretion of talinolol in human beings: a new type of drug/drug interaction. Clin Pharmacol Ther. 2000;68(4):345–355.
    1. Greiner B, et al. The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J Clin Investig. 1999;104(2):147–153.
    1. Gurley BJ, et al. Gauging the clinical significance of P-glycoprotein-mediated herb-drug interactions: comparative effects of St. John’s wort, Echinacea, clarithromycin, and rifampin on digoxin pharmacokinetics. Mol Nutr Food Res. 2008;52(7):772–779.
    1. Hamman MA, et al. The effect of rifampin administration on the disposition of fexofenadine. Clin Pharmacol Ther. 2001;69(3):114–121.
    1. Hartter S, et al. Decrease in the oral bioavailability of dabigatran etexilate after co-medication with rifampicin. Br J Clin Pharmacol. 2012;74(3):490–500.
    1. Lutz JD, et al. Cytochrome P450 3A induction predicts P-glycoprotein induction. Part 1: establishing induction relationships using ascending dose rifampin. Clin Pharmacol Ther. 2018;104(6):1182–1190.
    1. Rameis H. On the interaction between phenytoin and digoxin. Eur J Clin Pharmacol. 1985;29(1):49–53.
    1. Bergen DC. Pharmacokinetics of phenytoin: reminders and discoveries. Epilepsy Curr. 2009;9(4):102–104.
    1. Ahn JE, et al. Phenytoin half-life and clearance during maintenance therapy in adults and elderly patients with epilepsy. Neurology. 2008;71(1):38–43.
    1. Wang H, et al. Human constitutive androstane receptor mediates induction of CYP2B6 gene expression by phenytoin. J Biol Chem. 2004;279(28):29295–29301.
    1. Strandjord RE, Johannessen SI, Halpern LM. One daily dose of diphenylhydantoin for patients with epilepsy. Epilepsia. 1974;15(3):317–327.
    1. Yamada S, et al. Effects of the P-glycoprotein inducer carbamazepine on fexofenadine pharmacokinetics. Ther Drug Monit. 2009;31(6):764–768.
    1. Lutz JD, et al. Cytochrome P450 3A induction predicts P-glycoprotein induction. Part 2: prediction of decreased substrate exposure after rifabutin or carbamazepine. Clin Pharmacol Ther. 2018;104(6):1191–1198.
    1. Giessmann T, et al. Carbamazepine regulates intestinal P-glycoprotein and multidrug resistance protein MRP2 and influences disposition of talinolol in humans. Clin Pharmacol Ther. 2004;76(3):192–200.
    1. Bertilsson L, Tomson T. Clinical pharmacokinetics and pharmacological effects of carbamazepine and carbamazepine-10,11-epoxide. An update. Clin Pharmacokinet. 1986;11(3):177–198.
    1. Kuhnz W, et al. Carbamazepine and carbamazepine-10,11-epoxide during pregnancy and postnatal period in epileptic mother and their nursed infants: pharmacokinetics and clinical effects. Pediatr Pharmacol (New York) 1983;3(3–4):199–208.
    1. Bertilsson L. Clinical pharmacokinetics of carbamazepine. Clin Pharmacokinet. 1978;3(2):128–143.
    1. Chan KK, et al. Bioequivalence of carbamazepine chewable and conventional tablets: single-dose and steady-state studies. J Pharm Sci. 1985;74(8):866–870.
    1. Faucette SR, et al. Relative activation of human pregnane X receptor versus constitutive androstane receptor defines distinct classes of CYP2B6 and CYP3A4 inducers. J Pharmacol Exp Ther. 2007;320(1):72–80.
    1. Dresser GK, et al. Coordinate induction of both cytochrome P4503A and MDR1 by St John’s wort in healthy subjects. Clin Pharmacol Ther. 2003;73(1):41–50.
    1. Mueller SC, et al. Effect of St John’s wort dose and preparations on the pharmacokinetics of digoxin. Clin Pharmacol Ther. 2004;75(6):546–557.
    1. Schulz HU, et al. Investigation of pharmacokinetic data of hypericin, pseudohypericin, hyperforin and the flavonoids quercetin and isorhamnetin revealed from single and multiple oral dose studies with a hypericum extract containing tablet in healthy male volunteers. Arzneimittelforschung. 2005;55(10):561–568.
    1. Skinner MH, Blaschke TF. Clinical pharmacokinetics of rifabutin. Clin Pharmacokinet. 1995;28(2):115–125.
    1. Wang SY, et al. Effect of quercetin on P-glycoprotein transport ability in Chinese healthy subjects. Eur J Clin Nutr. 2013;67(4):390–394.
    1. Kim KA, Park PW, Park JY. Short-term effect of quercetin on the pharmacokinetics of fexofenadine, a substrate of P-glycoprotein, in healthy volunteers. Eur J Clin Pharmacol. 2009;65(6):609–614.
    1. Moon YJ, et al. Quercetin pharmacokinetics in humans. Biopharm Drug Dispos. 2008;29(4):205–217.
    1. Li L, et al. Bioactive terpenoids and flavonoids from Ginkgo biloba extract induce the expression of hepatic drug-metabolizing enzymes through pregnane X receptor, constitutive androstane receptor, and aryl hydrocarbon receptor-mediated pathways. Pharm Res. 2009;26(4):872–882.
    1. He X, et al. Effects of curcumin on the pharmacokinetics of talinolol in human with ABCB1 polymorphism. Xenobiotica. 2012;42(12):1248–1254.
    1. Juan H, Jing T, Wan-Hua Y, Juan S, Xiao-Lei L, Wen-Xing P. P-gp induction by curcumin: an effective antidotal pathway. J Bioequivalence Bioavailab. 2013;5(6):236–241.
    1. Cheng D, et al. Pharmacokinetics, pharmacodynamics, and PKPD modeling of curcumin in regulating antioxidant and epigenetic gene expression in healthy human volunteers. Mol Pharm. 2019;16(5):1881–1889.
    1. Kluth D, et al. Modulation of pregnane X receptor- and electrophile responsive element-mediated gene expression by dietary polyphenolic compounds. Free Radic Biol Med. 2007;42(3):315–325.
    1. Yamazaki S, Costales C, Lazzaro S, Eatemadpour S, Kimoto E, Varma MV. PBPK modeling approach to predict rifampin-mediated intestinal P-glycoprotein induction. Clin Pharmacol Ther Pharmacom Syst Pharmacol. 2019;8(9):634–642.
    1. Iisalo E. Clinical pharmacokinetics of digoxin. Clin Pharmacokinet. 1977;2(1):1–16.
    1. Klotz U, Antonin KH. Biliary excretion studies with digoxin in man. Int J Clin Pharmacol Biopharm. 1977;15(7):332–334.
    1. Caldwell JH, Cline CT. Biliary excretion of digoxin in man. Clin Pharmacol Ther. 1976;19(4):410–415.
    1. Magnusson MO, et al. Pharmacodynamics of carbamazepine-mediated induction of CYP3A4, CYP1A2, and Pgp as assessed by probe substrates midazolam, caffeine, and digoxin. Clin Pharmacol Ther. 2008;84(1):52–62.
    1. Nishimura M, Naito S, Yokoi T. Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies. Drug Metab Pharmacokinet. 2004;19(2):135–149.
    1. Oscarson M, et al. Transcriptional profiling of genes induced in the livers of patients treated with carbamazepine. Clin Pharmacol Ther. 2006;80(5):440–456.
    1. Schwarz UI, et al. Induction of intestinal P-glycoprotein by St John’s wort reduces the oral bioavailability of talinolol. Clin Pharmacol Ther. 2007;81(5):669–678.
    1. Chrubasik-Hausmann S, Vlachojannis J, McLachlan AJ. Understanding drug interactions with St. John’s wort (Hypericum perforatum L.): impact of hyperforin content. J Pharm Pharmacol. 2019;71(1):129–138.
    1. Yu CP, et al. Quercetin and rutin reduced the bioavailability of cyclosporine from Neoral, an immunosuppressant, through activating P-glycoprotein and CYP 3A4. J Agric Food Chem. 2011;59(9):4644–4648.
    1. Wang YH, et al. Lethal quercetin–digoxin interaction in pigs. Life Sci. 2004;74(10):1191–1197.
    1. Ha HR, et al. In vitro inhibition of midazolam and quinidine metabolism by flavonoids. Eur J Clin Pharmacol. 1995;48(5):367–371.
    1. Elbarbry F, Ung A, Abdelkawy K. Studying the inhibitory effect of quercetin and thymoquinone on human cytochrome P450 enzyme activities. Pharmacogn Mag. 2018;13(Suppl 4):S895–S899.
    1. Raucy JL. Regulation of CYP3A4 expression in human hepatocytes by pharmaceuticals and natural products. Drug Metab Dispos. 2003;31(5):533–539.
    1. Mitsunaga Y, et al. Effect of bioflavonoids on vincristine transport across blood–brain barrier. Eur J Pharmacol. 2000;395(3):193–201.
    1. Ravikumar Reddy D, et al. Natural flavonoids silymarin and quercetin improve the brain distribution of co-administered P-gp substrate drugs. Springerplus. 2016;5(1):1618.
    1. Zhang W, Lim LY. Effects of spice constituents on P-glycoprotein-mediated transport and CYP3A4-mediated metabolism in vitro. Drug Metab Dispos. 2008;36(7):1283–1290.
    1. Kusuhara H, et al. Pharmacokinetic interaction study of sulphasalazine in healthy subjects and the impact of curcumin as an in vivo inhibitor of BCRP. Br J Pharmacol. 2012;166(6):1793–1803.
    1. Xiao CQ, et al. Effect of genistein on the activities of cytochrome P450 3A and P-glycoprotein in Chinese healthy participants. Xenobiotica. 2012;42(2):173–178.
    1. Ellis LC, Hawksworth GM, Weaver RJ. ATP-dependent transport of statins by human and rat MRP2/Mrp2. Toxicol Appl Pharmacol. 2013;269(2):187–194.
    1. Niemi M, et al. Association of genetic polymorphism in ABCC2 with hepatic multidrug resistance-associated protein 2 expression and pravastatin pharmacokinetics. Pharmacogenet Genomics. 2006;16(11):801–808.
    1. Fromm MF, et al. The effect of rifampin treatment on intestinal expression of human MRP transporters. Am J Pathol. 2000;157(5):1575–1580.
    1. Marschall HU, et al. Complementary stimulation of hepatobiliary transport and detoxification systems by rifampicin and ursodeoxycholic acid in humans. Gastroenterology. 2005;129(2):476–485.
    1. Zhang W, et al. Role of BCRP 421C>A polymorphism on rosuvastatin pharmacokinetics in healthy Chinese males. Clin Chim Acta. 2006;373(1–2):99–103.
    1. Ho RH, et al. Effect of drug transporter genotypes on pravastatin disposition in European- and African-American participants. Pharmacogenet Genomics. 2007;17(8):647–656.
    1. Yu Y, et al. Population pharmacokinetics of talazoparib in patients with advanced cancer. J Clin Pharmacol. 2020;60:218–228.
    1. Nademanee K, et al. Amiodarone and thyroid function. Prog Cardiovasc Dis. 1989;31(6):427–437.
    1. Su SF, Huang JD. Inhibition of the intestinal digoxin absorption and exsorption by quinidine. Drug Metab Dispos. 1996;24(2):142–147.
    1. Calvo MV, et al. Interaction between digoxin and propafenone. Ther Drug Monit. 1989;11(1):10–15.
    1. Backman JT, et al. Concentrations and effects of oral midazolam are greatly reduced in patients treated with carbamazepine or phenytoin. Epilepsia. 1996;37(3):253–257.
    1. Ucar M, et al. Carbamazepine markedly reduces serum concentrations of simvastatin and simvastatin acid. Eur J Clin Pharmacol. 2004;59(12):879–882.
    1. Lee SD, Osei-Twum JA, Wasan KM. Dose-dependent targeted suppression of P-glycoprotein expression and function in Caco-2 cells. Mol Pharm. 2013;10(6):2323–2330.
    1. European Medicines Agency, Committee for Medicinal Products for Human Use. Talazoparib (Talzenna) Assessment Report; 2019. p. 58.
    1. Rostami-Hodjegan A. Physiologically based pharmacokinetics joined with in vitro-in vivo extrapolation of ADME: a marriage under the arch of systems pharmacology. Clin Pharmacol Ther. 2012;92(1):50–61.
    1. Pan Y, et al. The application of physiologically based pharmacokinetic modeling to predict the role of drug transporters: scientific and regulatory perspectives. J Clin Pharmacol. 2016;56(Suppl 7):S122–S131.
    1. Tubic M, et al. In silico modeling of non-linear drug absorption for the P-gp substrate talinolol and of consequences for the resulting pharmacodynamic effect. Pharm Res. 2006;23(8):1712–1720.

Source: PubMed

3
Abonnieren