Elsevier

Neuropharmacology

Volume 55, Issue 8, December 2008, Pages 1364-1375
Neuropharmacology

Several major antiepileptic drugs are substrates for human P-glycoprotein

https://doi.org/10.1016/j.neuropharm.2008.08.032Get rights and content

Abstract

One of the current hypotheses of pharmacoresistant epilepsy proposes that transport of antiepileptic drugs (AEDs) by drug efflux transporters such as P-glycoprotein (Pgp) at the blood–brain barrier may play a significant role in pharmacoresistance in epilepsy by extruding AEDs from their intended site of action. However, several recent in vitro studies using cell lines that overexpress efflux transporters indicate that human Pgp may not transport AEDs to any relevant extent. In this respect it has to be considered that most AEDs are highly permeable, so that conventional bi-directional transport assays as used in these previous studies may fail to identify AEDs as Pgp substrates, particularly if these drugs are not high-affinity substrates for Pgp. In the present study, we used a modified transport assay that allows evaluating active transport independently of the passive permeability component. In this concentration equilibrium transport assay (CETA), the drug is initially added at identical concentration to both sides of a polarized, Pgp-overexpressing cell monolayer instead of applying the drug to either the apical or basolateral side for studying bi-directional transport. Direct comparison of the conventional bi-directional (concentration gradient) assay with the CETA, using MDR1-transfected LLC cells, demonstrated that CETA, but not the conventional assay, identified phenytoin and phenobarbital as substrates of human Pgp. Furthermore, directional transport was determined for lamotrigine and levetiracetam, but not carbamazepine. Transport of AEDs could be completely or partially (>50%) inhibited by the selective Pgp inhibitor, tariquidar. However, transport of phenobarbital and levetiracetam was also inhibited by MK571, which preferentially blocks transport by multidrug resistance transporters (MRPs), indicating that, in addition to Pgp, these AEDs are substrates of MRPs. The present study provides the first direct evidence that several AEDS are substrates of human Pgp, thus further substantiating the transporter hypothesis of pharmacoresistant epilepsy.

Introduction

Resistance to antiepileptic drugs (AEDs) is a major, unresolved problem in epilepsy therapy, affecting about 30–40% of all patients (Kwan and Brodie, 2000, Chang and Lowenstein, 2003). Most patients with AED-resistant epilepsy are resistant to several, if not all, AEDs, despite the fact that these drugs act by different mechanisms. The consequences of uncontrolled epilepsy can be severe, and include shortened lifespan, bodily injury, neuropsychologial and psychiatric impairment, and social disability (Sperling, 2004). Consequently, there is a pressing need to develop new and more effective treatment strategies to counteract or prevent pharmacoresistance. For this goal, we need to understand the mechanisms underlying AED resistance. One of the candidate mechanisms that has attracted growing interest is the limitation of AED access to epileptogenic brain region(s) by localized overexpression of drug efflux transporters such as P-glycoprotein (Pgp) at the blood–brain barrier (BBB) (Löscher and Potschka, 2005a). A prerequisite for this transporter hypothesis of drug resistance is that AEDs are substrates of human Pgp.

However, several recent reports, including studies by our group, have indicated that, in contrast to rodent Pgp, human Pgp may not transport AEDs to any relevant extent (Schinkel et al., 1996, Mahar Doan et al., 2002, Crowe and Teoh, 2006, Baltes et al., 2007a, Baltes et al., 2007b). These recent reports have used conventional (bi-directional) transport assays with polarized intestinal (Caco-2) or kidney (MDCKII, LLC) cell lines expressing the human multidrug resistance-1 (MDR1; ABCB1) gene that encodes Pgp. The transport assays were performed in a conventional manner with the Transwell® system that allows studying drug transport between an apical and basolateral compartment separated by a polarized cell monolayer on a polyester filter membrane, applying the AED to either the apical or basolateral chamber for studying bi-directional transport. However, because most AEDs are very lipophilic, passive transcellular diffusion could form a bias in such assays by concealing active transport. Thus, the conventional bi-directional transport assay may fail to identify highly permeable compounds as Pgp substrates, particularly if they are not high-affinity substrates for this efflux transporter (FDA, 2006). In line with this possibility, Robey et al. (2008) recently suggested that it may be that AEDs are Pgp substrates but are not so well transported that they can be detected by the model systems used in previous studies.

This prompted us to modify the transport assay in a way that allows evaluating active transport independently of the passive permeability component. For this purpose, we adapted a method recently described for measuring Pgp-mediated transport of highly permeable antibiotics in the Caco-2 model (Pachot et al., 2003). Instead of applying the drug to either the apical or basolateral chamber for studying bi-directional transport, the drug is initially added at identical concentration to both chambers, resulting in concentration equilibrium conditions (Pachot et al., 2003). This concentration equilibrium transport assay thus minimizes the problem of drug concentration gradients that is known to affect identification of highly permeable compounds as Pgp substrates. In the present study, the concentration equilibrium transport assay was used to determine whether major AEDs are transported by human Pgp, using kidney cell lines transfected with MDR1. The known high-affinity Pgp substrates vinblastine and digoxin were included in the study for comparison.

Section snippets

Cell lines and cell cultures

LLC-PK1 cells transfected with human MDR1 (LLC–MDR1) and respective wildtype (Wt) LLC cells were kindly provided by Prof. P. Borst (The Netherlands Cancer Institute, Amsterdam, Netherlands). Some experiments were also performed with MDCK type II cells transfected with human MDR1 (MDCKII–MDR1) and respective wildtype cells, which were also kindly provided by Prof. P. Borst. After obtaining the cells, they were cultured as described in detail recently (Baltes et al., 2007a, Baltes et al., 2007b).

Comparison of transport of vinblastine, digoxin, phenytoin and phenobarbital by Pgp in conventional vs. concentration equilibrium transport assays

Under concentration gradient conditions as commonly used in transcellular transport assays in Pgp-overexpressing cell lines, the prototype Pgp substrates vinblastine and digoxin exhibited directional (basolateral to apical) transport with cTRs of 3.01 and 3.41, respectively, in LLC–MDR1 cells (Fig. 2). With digoxin, directional transport was also observed in LLC wildtype cells, indicating transport by endogenous pig Pgp. Substantial basolateral to apical transport in wildtype cells was also

Discussion

Our recent study (Baltes et al., 2007a) indicating species differences in the Pgp-mediated transport of AEDs with significant transport of phenytoin and levetiracetam by mouse but not human Pgp in transfected LLC cells has cast serious doubt on the hypothesis that overexpression of multidrug transporters such as Pgp may mediate resistance to AEDs in patients with epilepsy (Löscher and Sills, 2007). The transporter hypothesis of AED-resistant epilepsy is fundamentally supported by four key

Acknowledgements

We thank Dr. Astrid Volz (Boehringer Ingelheim, Department of Drug Discovery Support, Biberach, Germany) for discussions on the concentration equilibrium transport assay and Prof. Piet Borst (The Netherlands Cancer Institute) and his group for kindly providing us with the cell lines used in this study and the information that Prof. Borst and Dr. A.H. Schinkel have previously used the concentration equilibrium transport assay for identifying transport by Pgp. The skilful technical assistance of

References (70)

  • H. Potschka et al.

    Inhibition of multidrug transporters by verapamil or probenecid does not alter blood–brain barrier penetration of levetiracetam in rats

    Epilepsy Res.

    (2004)
  • S. Santaguida et al.

    Side by side comparison between dynamic versus static models of blood–brain barrier in vitro: a permeability study

    Brain Res.

    (2006)
  • A.H. Schinkel et al.

    Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview

    Adv. Drug Deliv. Rev.

    (2003)
  • M.E. Taub et al.

    Optimized conditions for MDCK permeability and turbidimetric solubility studies using compounds representative of BCS classes I–IV

    Eur. J. Pharm. Sci.

    (2002)
  • Z.H. Yang et al.

    P-glycoprotein-mediated efflux of phenobarbital at the blood–brain barrier evidence from transport experiments in vitro

    Epilepsy Res.

    (2008)
  • C.J. Bachmeier et al.

    Comparison of drug efflux transport kinetics in various blood–brain barrier models

    Drug Metab. Dispos.

    (2006)
  • S. Baltes et al.

    All-trans retinoic acid enhances differentiation and influences permeability of intestinal Caco-2 cells under serum-free conditions

    Dev. Growth Differ.

    (2004)
  • S. Baltes et al.

    Valproic acid is not a substrate for P-glycoprotein or multidrug resistance proteins 1 and 2 in a number of in vitro and in vivo transport assays

    J. Pharmacol. Exp. Ther.

    (2007)
  • J.C. Baron et al.

    Brain regional pharmacokinetics of 11C-labeled diphenylhydantoin: positron emission tomography in humans

    Neurology

    (1983)
  • P. Borst et al.

    A family of drug transporters: the multidrug resistance-associated proteins

    J. Natl. Cancer Inst.

    (2000)
  • D. Brügger et al.

    Induction of drug resistance and protein kinase C genes in A2780 ovarian cancer cells after incubation with antineoplastic agents at sublethal concentrations

    Anticancer Res.

    (2002)
  • B.S. Chang et al.

    Epilepsy

    N. Engl. J. Med.

    (2003)
  • A. Crowe et al.

    Limited P-glycoprotein mediated efflux for anti-epileptic drugs

    J. Drug Target

    (2006)
  • L. Cucullo et al.

    Development of a humanized in vitro blood–brain barrier model to screen for brain penetration of antiepileptic drugs

    Epilepsia

    (2007)
  • R. Evers et al.

    Drug export activity of the human canalicular multispecific organic anion transporter in polarized kidney MDCK cells expressing cMOAT (MRP2) cDNA

    J. Clin. Invest.

    (1998)
  • S. Eyal et al.

    The antiepileptic and anticancer agent, valproic acid, induces P-glycoprotein in human tumour cell lines and in rat liver

    Br. J. Pharmacol.

    (2006)
  • S.D. Flanagan et al.

    Comparison of furosemide and vinblastine secretion from cell lines overexpressing multidrug resistance protein (P-glycoprotein) and multidrug resistance-associated proteins (MRP1 and MRP2)

    Pharmacology

    (2002)
  • Food and Drug Administration (FDA) and U.S. Department of Health and Human Services

    Guidance for industry. Drug interaction studies – study design, data analysis, and implications for dosing and labeling

  • T. Giessmann et al.

    Carbamazepine regulates intestinal P-glycoprotein and multidrug resistance protein MRP2 and influences disposition of talinolol in humans

    Clin. Pharmacol. Ther.

    (2004)
  • C.C. Hung et al.

    Functional evaluation of polymorphisms in the human ABCB1 gene and the impact on clinical responses of antiepileptic drugs

    Pharmacogenet. Genom.

    (2008)
  • Y. Kimura et al.

    Mechanism of multidrug recognition by MDR1/ABCB1

    Cancer Sci.

    (2007)
  • P. Kwan et al.

    Early identification of refractory epilepsy

    N. Engl. J. Med.

    (2000)
  • P. Kwan et al.

    Potential role of drug transporters in the pathogenesis of medically intractable epilepsy

    Epilepsia

    (2005)
  • O. Langer et al.

    Pharmacoresistance in epilepsy: a pilot PET study with the P-glycoprotein substrate R-[C]verapamil

    Epilepsia

    (2007)
  • W. Löscher et al.

    Kinetics of penetration of common antiepileptics drugs into cerebrospinal fluid

    Epilepsia

    (1984)
  • Cited by (0)

    View full text