• Nie Znaleziono Wyników

The multidrug transporter P-glycoprotein in pharmacoresistance to antiepileptic drugs

N/A
N/A
Protected

Academic year: 2022

Share "The multidrug transporter P-glycoprotein in pharmacoresistance to antiepileptic drugs"

Copied!
9
0
0

Pełen tekst

(1)

Review

The multidrug transporter P-glycoprotein in pharmacoresistance to antiepileptic drugs

Karolina M. Stêpieñ1, Micha³ Tomaszewski2,3, Joanna Tomaszewska3,4, Stanis³aw J. Czuczwar5,6

1Clinical Biochemistry and Metabolic Medicine Department, Central Manchester Foundation Trust, Oxford Road, M13 9WL Manchester, UK

2Department of Cardiology,3Chair of Internal Medicine and Department of Internal Medicine in Nursing, Medical University of Lublin, Jaczewskiego 8, PL 20-954 Lublin, Poland

4Department of Vitreoretinal Surgery, Medical University of Lublin, Chmielna 1, PL 20-079 Lublin, Poland

5Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8, PL 20-090 Lublin, Poland

6Department of Physiopathology, Institute of Agricultural Medicine, Jaczewskiego 2, PL 20-950 Lublin, Poland Correspondence: Micha³ Tomaszewski, e-mail: mdtomaszewski@yahoo.com

Abstract:

This review provides an overview of the knowledge on P-glycoprotein (P-gp) and its role as a membrane transporter in drug resis- tance in epilepsy and drug interactions. Overexpression of P-gp, encoded by the ABCB1 gene, is involved in resistance to antiepilep- tic drugs (AEDs), limits gastrointestinal absorption and brain access of AEDs. Although several association studies on ABCB1 gene with drug disposition and disease susceptibility are completed to date, the data remain unclear and incongruous. Although the litera- ture describes other multidrug resistance transporters, P-gp is the main extensively studied drug efflux transporter in epilepsy.

Key words:

P-glycoprotein, multidrug transporter, pharmacoresistance, antiepileptic drugs

Abbreviations: AEDs – antiepileptic drugs, ATP – adenosine triphosphate, BBB – blood brain barrier, BCRP – breast cancer resistance protein, DRE – drug resistant epilepsy, MRPs – mul- tidrug resistance-associated proteins, P-gp – P-glycoprotein, PKA – protein kinase A, PKC – protein kinase C, SNP – single nucleotide polymorphism, TLE – temporal lobe epilepsy, TNF – tumor necrosis factor

Introduction

Epilepsy is a chronic and often progressive brain disorder, characterized by the periodic and unpredictable occurrence

of seizures. It affects approximately 1 to 2% [35, 51]

of the population. The main treatment of choice in epilepsy is chronic administration of antiepileptic drugs (AEDs) [10]. However, seizures continue to re- cur in 30% of people with epilepsy despite drug ther- apy. Current treatment options are limited among these patients. Morbidity and mortality are increased as well [64]. Long periods of drug resistant epilepsy (DRE) are associated with difficulties in psychosocial functions such as dependent behavior, lifestyle re- strictions, poor academic achievement and decline in self-esteem [15, 38]. Intractable epilepsy is associated with five-fold higher mortality rate when compared to

Pharmacological Reports 2012, 64, 1011–1019 ISSN 1734-1140

Copyright © 2012 by Institute of Pharmacology Polish Academy of Sciences

(2)

Drug-resistant epilepsy (DRE)

The definition of DRE is elusive. A number of plausi- ble hypotheses of DRE have been proposed. Most cli- nicians would consider an epilepsy pharmacoresistant that had not been controlled by any of two to three first-line AEDs usually used for a given epilepsy syn- drome [42]. Potential causes of apparent resistance – such as misdiagnosis, non-compliance, inappropriate treatment or interaction between AEDs must be ex- cluded [33, 64]. Failure of drug response is a major limitation in the treatment of epilepsy.

The epilepsy itself has multiple etiologies so the intractability to AEDs may also be multifactorial. It has not been conclusively determined whether phar- macoresistance develops as the result of the disease process or exists at the time of initial seizure [15].

Elger [15] suggests that drug resistance is present in the early stage of the disease. Patients who do not respond to the first AED have a chance of only 10%

or lower to be controlled by other AEDs (even those

suffer from partial seizures [56].

The pathophysiological basis of pharmacoresis- tance in epilepsy is still poorly understood. One of suggested mechanisms refers to reduced target sensitivity and may occur through alterations of the composition and functionality of voltage-gated ion channels or neurotransmitter receptors in epileptogenic brain tissue. Another DRE hypothesis refers to P-glycoprotein (P-gp) expressed at the blood brain barrier (BBB) as a result of seizures, induction mediated by the nuclear receptor PXR, or genetic factors.

This review will present the overview of the current knowledge on biochemical properties and mechanisms of P-gp, particularly that several AEDs frequently used in the treatment of epilepsies are substrates of P-gp both in rodents (gabapentin, lamotrigine, phenobarbi- tal, phenytoin and topiramate) and humans (phenytoin, phenobarbital, lamotrigine and levetiracetam) [4, 41, 44] (Tab. 1). Data on carbamazepine, levetiracetam and valproic acid are not clear [44, 48, 77].

Biochemical and functional properties of P-pg

P-gp is a prototype of membrane transporters. It is a 170-kD transmembrane glycoprotein that contains 1280 amino acids. It is a subject of several post- translational modifications, such as N-glycosylation, phosphorylation and ubiquitination [79] and consists of two similar halves of 610 amino acids each, joined by a linker region consisting of 60 charged residues.

Each of the two halves forms six transmembrane spans and hydrolyzes ATP during molecular transport.

The presence of both halves is essential for the trans- port activity [79].

According to Schmidt and Löscher [59], P-gp meets all the criteria for the transporter hypothesis as 1) it is increased in epileptogenic brain tissue of ro- dents; 2) associated with lower brain levels of AEDs;

3) higher in AED-resistant rats than in responsive ani- mals and 4) coadministration of the highly selective P-gp inhibitor, tariquidar, reverses AED resistance.

Thus, P-gp plays a significant role in mediating resis-

Tab. 1. Antiepileptic drugs (AEDs) that are substrates/inhibitors of P-glycoprotein with high/moderate or low/none affinity

AEDs References

Moderate/High affinity Low/No affinity Carbamazepine [6, 63, 73, 76, 77] [4, 22, 39, 43, 44,

52, 60]

Phenytoin [6, 14, 39, 41, 43, 44, 48, 63, 73, 75, 76]

Phenobarbital [8, 39, 41, 43, 44, 73, 75, 76]

Levetiracetam [6, 39, 41, 43, 44, 48, 76]

Lamotrigine [39, 41, 43, 44, 73, 76]

Gabapentin [73, 76]

Topiramate [48] [73, 76]

Valproate [4, 5, 39, 43, 44,

48, 73, 76]

(3)

tance to AEDs in rodent models of temporal lobe epi- lepsy (TLE) and inhibition of P-gp can outwit this mechanism [59].

P-gp is specifically localized to secretory surfaces of liver, pancreas, renal tubules, small intestine and bile canaliculi acting as an energy-dependent pump for liphophilic compounds, including noxious xenobi- otics [38, 57]. It is also present in the brain capillaries that form the BBB; localized in the lining endothe- lium where it can restrict entry of lipophilic drugs into the brain and export substances, such as b-amyloid, from neuroparenchyma [32, 34]. P-gp pumps xenobi- otics from intracellular space back to the capillary lumen, thereby maintains the integrity of BBB and reduces the cerebral accumulation of substrate drugs [32].

P-gp is the first discovered human ABC transporter in drug-resistance ovarian cells obtained from Chinese hamsters [25]. It was initially discovered in multidrug resistance of cancer cells, but was also the first drug ef- flux transporter in multidrug resistance that was de- tected in endothelial cells of human BBB [20, 47].

Two genes in humans encode P-gp: MDR1 (system- atic name ABCB1) [67] and MDR2 (ABCB4). In ro- dents, it is encoded by three genes mdr1a, mdr1b and mdr2 that have strikingly similar sequence to several bacterial transport proteins [19]. P-gp has very chemi- cally diverse substrates and inhibitors [31].

Overexpression of P-gp

Brain capillaries restrict the penetration of hydro- philic, polar and protein-bound compounds, whereas non-polar and highly lipid-soluble drugs penetrate easily through BBB by passive diffusion [42]. To penetrate BBB drugs have to be highly liphophilic.

Multidrug transporters are believed to function as an active defense mechanism, preventing lipophilic sub- stances from reaching the brain through either the BBB or blood-cerebrospinal fluid barrier [32, 66].

P-gp is believed to cause multidrug resistance by reducing intracellular drug accumulation through its function as an active efflux pump [31]. It prevents high accumulation of the drug in the brain tissue [69]. The active transport from the brain back into blood vessels causes lower brain accumulation of drugs than it would be expected based on their lipophilicity [69].

It has been hypothesized that overexpression of ef- flux transporters at the BBB [25], which prevents AEDs from reaching sufficiently high brain concen- trations despite adequate plasma levels, could be one such mechanism [31]. P-gp overexpression may be the consequence of (i) seizures themselves, (ii) PXR- mediated upregulation through exposure to certain an- ticonvulsants such as carbamazepine, or (iii) genetic variation [37].

Additionally, it has been shown that P-gp can be induced by AEDs in both endothelial and astroglial cells in culture [74]. On the contrary, Ambroziak et al.

[1] have demonstrated that exposure to major antiepileptic drugs (such as phenobarbital, phenytoin and carbamazepine) does not alter the functionality of P-gp [1, 4].

The expression of P-gp in different individuals var- ies probably due to genetic and environmental influ- ences [62]. It is crucial whether overexpression of ef- flux transporters in epileptic brain tissue is constitu- tive or acquired/induced or both mechanisms coexist.

A constitutive overexpression could be a result of ge- netic predisposition or it could be intrinsic to the de- velopment of the specific pathology [30, 31]. Theory that overexpression of efflux transporters may be con- stitutive or intrinsic, may be supported by the obser- vation of P-gp upregulation in patients’ post-mortem tissues with malformations of cortical development that had died before experiencing seizures or expo- sure to AEDs [67]. Kwan and Brodie [31] hypothe- size that increased expression of drug transporters is associated with refractory epilepsy and that AEDs are substrates of these transporters. The evidence for upregulation of efflux transporters such as P-gp was derived from brain tissues removed during epilepsy surgery from patients with drug resistant epilepsy.

Pathologically elevated expression of P-gp has been found in resected brain tissue of patients with refrac- tory temporal lobe epilepsy (TLE) undergoing epi- lepsy surgery as well as in limbic brain regions of mouse and rat models of TLE [38, 40, 41].

Recent report by Bankstahl et al. [7] has demon- strated in experimental study on mice that increased P-gp expression and activity post status epilepticus occurs not only in primary epileptogenic regions e.g., hippocampus, but also in the thalamus and the cere- bellum [7].

In the animal models, increased expression of P-gp was associated with decreased brain concentration of AEDs such as phenytoin [40, 41].

The multidrug transporter P-glycoprotein

Karolina M. Stêpieñ et al.

(4)

protein kinases. It has been documented that P-gp contains sites with structural features mimicking phos- phorylation sites for protein kinases such as protein kinase C (PKC) or protein kinase A (PKA) [12]. P-gp is also phosphorylated in vivo and the linker region (amino acids 629–686) was identified to be the major phospho- rylation domain [17]. It has been shown that substitution of PKA and PKC phosphorylatable residues in the P-gp linker region affects the stimulation of P-gp ATP-ase activity by some substrates [17].

Pharmacogenetics of P-gp

Genetic polymorphism in transporters may explain why two patients with the same type of epilepsy may have different response to AEDs [15]. Some AEDs are substrates [22] and inhibitors of P-gp (phenytoin, carbamazepine, lamotrigine, phenobarbital, valproic acid, levetiracetam, gabapentin) that can be affected by ABCB1 gene polymorphisms (Tab. 1). Because the efflux drug transporter P-gp is highly expressed at BBB, genetic variation in its expression or functional- ity could directly affect brain uptake and extrusion of AEDs. The overexpression of several ABC-trans- porters, particularly P-gp (MDR-1/ABCB1 gene) has been recognized to play a central role in the pharma- coresistant phenotype in epilepsy by limiting the drug efficacy as well as the plasmatic levels of AEDs in affected cases [36, 55].

There is still considerable controversy whether polymorphisms in the ABCB1 gene encoding P-gp could influence drug-response and seizure frequency [27, 61]. Recent genetic association studies have indicated an association of the 3435CC genotype, which is associated with increased P-gp expression, with AED resistant epilepsy [37]. Noteworthy, cerebrospinal fluid concentrations of phenobarbital were significantly lower in epilepsy patients with the 3435CC genotype [8]. Mosyagin et al. [49], however, examined the impact of polymorphisms 3435CT and 2677GT in the ABCB1 gene on the ABCB1 mRNA expression and P-gp content. Authors concluded that they cannot exclude an association of ABCB1 variants on P-gp function, but their results suggest that brain ABCB1 mRNA and protein expression is

phism (SNP), at the position C3435T in exon 26 of the MDR1 gene, is associated with differential intesti- nal expression of P-gp. However, the homozygous T-allele (T3435T or TT genotype) is associated with decreased MDR-1 expression by approximately 2-fold compared with patients homozygous for C-allele (C3435C or CC genotype), and heterozygous indi- viduals exhibit an intermediate phenotype (T3435C or CT genotype) [23, 35].

Importantly, the correlation between intestinal ex- pression of cytochrome P450 enzymes and ACB transporters with dose requirement and plasma levels of carbamazepine and phenytoin, evaluated in 44 epi- leptic patients, indicated that differences in intestinal MDR1 and MRP2 expression may influence carba- mazepine and phenytoin disposition and may account for interindividual pharmacokinetic variability [63].

Additionally, phenytoin plasma levels has been shown to be affected by polymorphisms in MDR1, CYP2C9 and CYP2C19 [26]. Results of a recent Japanese study showed an association between C3435T and low intestinal expression of cytochrome P450 (CYP3A4) that similarly to ABCB1 gene, is located at chromo- some 7q21 [18]. As a consequence, the induction of CYP3A4 is associated with MDR-1 induction [78].

The results of another recent study by Meng et al. [46]

were consistent with the published results for the Japanese patients and showed that the 3435TT geno- type was associated with increased P-gp expression [46] that was converse to the results of the study on Caucasian group of patients [23]. Authors concluded that patients with the 3435-TT genotype had signifi- cantly lower plasma carbamazepine concentrations than those with the 3435-CC genotype [46], but there were no significant associations between all the stud- ied genotypes, haplotypes and diplotypes involving SNPs of ABCB1 gene.

Several studies have been performed on patients treated with multiple AEDs. In studies performed on patients treated with monotherapy, an association between ABCB1_3435CT polymorphisms and drug- resistance was found in individuals treated with phenytoin [14] or phenobarbital [8], whereas in those with carbamazepine a reverse association [60] or non-association [52] was found.

The clinical impact of ABCB1 polymorphisms on AED resistance in patients with epilepsy has been

(5)

assessed in clinical studies with P-gp inhibitors, such as nimodipine or cyclooxygenase-2 inhibitors that significantly improve the anticonvulsive action of AEDs [24, 71]. These drugs optimize the control of P-gp expression, improve AEDs brain penetration, help overcome the pharmacoresistance and control seizures [18]. One study examining an inhibitory interaction of several AEDs with P-gp in vitro at concentrations exceeding therapeutic plasma concentrations, has suggested that modulation of P-gp is not a major determinant of AED action in monotherapy. It is possible that additive or even synergistic inhibition of transport will occur when several AEDs that compete for P-gp binding sites are given concomitantly [73].

A meta-analysis by Nurmohamed et al. [51] has shown that although the ABCB1 gene had been highly investi- gated and plays many important roles, it is unlikely that the polymorphism at 3435 plays a major role in the devel- opment of drug resistance to AEDs [55]. Similarly, Haerian et al. [21] have failed to show an association be- tween the C3435T polymorphism in ABCB1 gene and the risk of drug-resistance [21]. Authors of meta-analyses do not recommend testing for C3435T polymorphisms of ABCB1 gene as the investigation will not yield any infor- mation that would be useful in the diagnosis, manage- ment or prognosis of epilepsy [9, 21].

The role of ABCC2 (MRP2) transporter is not well understood yet [68]. However, ABCC2 expression was reported in brain-derived endothelial cells from patients with drug-refractory epilepsy, indicating

a potential upregulation of ABCC2 because of repeti- tive seizures. This study has demonstrated a higher risk of AED failure in ABCC2 (MRP2)-24T allele carriers, possibly though a compensatory upregulation of ABCB1 (P-gp) [68].

P-gp vs. other drug transporters

P-gp compared to other known multidrug transporters has one known isoform, whereas multidrug resistance- associated proteins (MRPs) have multiple confirmed ones such as MRP1, MRP2, MRP3, MRP4 and MRP5 that were recently isolated in brain microvessel endo- thelial capillaries and on the luminal side of brain cap- illary endothelial cells [50, 72] (Tab. 2). Both P-gp and MRP have been also found to be expressed in TLE specimens [2, 66].

Breast cancer resistance protein (BCRP) also known as ABCG2/MXR/ABCP, is another ABC transporter family and expresses significant overlap in substrate specificity profile with P-gp [53]. BCRP seems to have a low affinity for AEDs so that its role in drug resistance epilepsy is not as important as it is for P-gp [13]. BCRP has been expressed in capillary endothelial cells, but in contrast to P-gp and MRP, BCRP levels are not changed in the brain tissue from patients with hippocampal sclerosis [2, 3]. Addition- ally, all three types of ABC transporters are induced

The multidrug transporter P-glycoprotein

Karolina M. Stêpieñ et al.

Tab. 2. Comparison of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP)

P-gp MRP References

Number of isoforms One More than one [50, 53, 72]

Hippocampal sclerosis expression Yes Yes [2, 3]

Seizure induced Yes Yes [70]

Transporter for AEDs Yes No [39, 43, 44, 77]

Effect on expression TNFa

IL-1b IL-6

Increased Increased Decreased

Increased No effect No effect

[29, 58]

[58]

[58]

Blood cells expressing transporter:

Erythrocytes Platelets Lymphocytes

Yes Yes Yes

Yes Yes No

[28]

[28]

[28]

(6)

frequency [70].

Interestingly, P-gp has been shown to be a trans- porter for major AEDs such as carbamazepine [77], phenytoin, lamotrigine, levetiracetam and phenobar- bital, however, MRP1, 2 and 5 has not been demon- strated to have such properties [39, 43, 44]. Zhang et al. [75] have exhibited that human P-gp is involved in concentration dependent transport of phenytoin and phenobarbital (but not ethosuximide) [75]. It is not known yet if BCRP is a transporter for AEDs.

Some studies examined the effect of inflammatory mediators on expression of multidrug transporters and have shown that the expression of P-gp can be in- creased by TNFa, but not by IL-1b or IL6 [58]. Simi- larly, MRP1 expression can be increased by TNFa and polyinosinic-cytidylic acid a ligand for Toll-like receptor-3 [29]. These results suggest that P-gp- expressing astrocytes in the close proximity to blood vessels form a glial barrier that may alter the ability of AEDs to access the epileptogenic focus. In addition, this barrier remains under the control of pro- inflammatory pathways [29]. Currently, there are no similar studies on BCRP and inflammatory mediators.

Multidrug transporters have been found to be pres- ent in glial cells in the brain, hippocampus, frontal and peripheral cortex [4, 65], but there is some evi- dence that MRP 1–5 may be expressed in blood cells such as erythocytes, platelets, and P-gp additionally in lymphocytes [28] (Tab. 2). The fact is important in the aspect of the transport of AEDs such as valproate. It has been suggested that valproate accumulates in erythrocytes when, as a result of drug transporters in- hibition at the erythrocyte membrane, its plasma con- centration significantly decreases during co-medica- tion with carbapenem antibiotics [45]. The reduction of valproate plasma levels was associated with the oc- currence of seizures in patients with epilepsy [16].

The transporter for valproate has not been identified as yet as there is no evidence that valproate is trans- ported by P-gp or MRPs [39, 43, 44].

Conclusion

In conclusion, the knowledge on pharmacoresistance mechanisms to AEDs becomes important in drug de-

nation with AEDs [18]. Finally, the identification of genetic polymorphisms, in either transporter or AEDs genes, would increase the chance of the antiepileptic therapy. However, the most recent data suggest that ABCB1 variants provide no evidence of an associa- tion of drug responsiveness to AEDs.

In view of current controversial knowledge about associations of polymorphisms and AEDs therapeutic efficacy, but also which AEDs are substrates for P-gp, further research is necessary to determine the sub- strate characteristics of different AEDs. It would be desirable to design an AED that would have low or medium affinity for multidrug transporter proteins.

AEDs are highly lipophilic agents and pass mem- branes by rapid diffusion. In addition, they generally pass the blood-brain barrier, and are only affected when pathophysiologic mechanisms upregulate P-gp- expression brain-penetration rates [54]. Modulating P-gp is likely to overcome resistance to some of AEDs, but definitely will not solve the issue of mul- tidrug resistance. Some advancement in the form of P-gp modulators such as cyclooxygenase-2 has been exhibited and requires further evaluation of its clinical relevance.

References:

1.Ambroziak K, Kuteykin-Teplyakov K, Luna-Tortos C, Al-Falah M, Fedrowitz M, Löscher W: Exposure to antiepileptic drugs does not alter the functionality of P-glycoprotein in brain capillary endothelial and kidney cell lines. Eur J Pharmacol, 2010, 628, 57–66.

2.Aronica E, Gorter JA, Ramkema S, Redeker S, Ozbas- Gerceker F, van Vliet EA, Scheffer GL et al.: Expression and cellular distribution of multidrug resistance-related proteins in the hippocampus of patients with mesial tem- poral lobe epilepsy. Epilepsia, 2004, 45, 441–451.

3.Aronica E, Gorter JA, Redeker S, van Vliet EA, Ramkema M, Scheffer GL, Scheper RJ et al.: Localiza- tion of breast cancer resistance protein (BCRP) in microvessel endothelium of human control and epileptic brain. Epilepsia, 2005, 46, 846–857.

4.Aronica E, Sisodiya SM. Gorter JA: Cerebral expression of drug transporters in epilepsy. Adv Drug Deliv Rev, 2012, 64, 919–929.

5.Baltes S, Fedrowicz M, Tortos CL, Potschka H, Löscher W: Valproic acid is not a substrate for P-glycoprotein or multidrug resistance proteins 1 and 2 in a number of in

(7)

vitro and in vivo transport assays. J Pharmacol Exp Ther, 2007, 320, 331–343.

6.Baltes S, Gastens AM, Fedrowicz M, Potschka V, Kaever V, Löscher W: Differences in the transport of the antiepileptic drugs phenytoin, levetiracetam and carba- mazepine by human and mouse P-glycoprotein. Neuro- pharmacology, 2007, 52, 333–346.

7.Bankstahl JP, Bankstahl M, Kuntner C, Stanek J, Wanek T, Meier M, Xiao-Qi D et al.: A novel positron emission tomography imaging protocol identifies seizure-induced regional overactivity of P-glycoprotein at the brain-brain barrier. J Neurosci, 2011, 31, 8803–8811.

8.Basic S, Hajnsek S, Bozina N, Filipcic I, Sporis D, Mislov D, Posavec A: The influence of C3435T poly- morphism of ABCB1 gene on penetration of phenobarbi- tal across blood-brain barrier in patients wit generalized epilepsy. Seizure, 2008, 17, 524–530.

9.Bournissen FG, Moretti ME, Juurlink DN, Koren G, Walker M, Finkelstein Y: Polymorphism of the MDR1/ABCB1 C3435T drug-transporter and resistance to anticonvulsant drugs: a meta-analysis. Epilepsia, 2009, 50, 898–903.

10.Browne TR, Holmes GL: Epilepsy. N Eng J Med, 2001, 344, 1145–1151.

11.Buck D, Baker GA, Jacoby A, Smith DF, Chadwick DW:

Patients’ experiences of injury as a result of epilepsy.

Epilepsia, 1997, 38, 439–444.

12.Chambers TC, Pohl J, Glass DB, Kuo JF: Phosphoryla- tion by protein kinase C and cyclic AMP-dependent pro- tein kinase od synthetic peptides derived from the linker region of human P-glycoprotein. Biochem J, 1994, 299, 309–315.

13.Cerveny L, Pavek P, Malakova J, Staud F, Fendrich Z:

Lack of interactions between breast cancer resistance protein (BCRP/ABCG22) and selected antiepileptic agents. Epilepsia, 2006, 47, 461–468.

14.Ebid AH, Ahmed MM, Mohammed SA: Therapeutic drug monitoring and clinical outcomes in epileptic Egyp- tian patients: a gene polymorphism perspective study.

Ther Drug Monit, 2007, 29, 305–312.

15.Elger CE: Pharmacoresistance: Modern concept and ba- sic data derived from human brain tissue. Epilepsia, 2003, 44, 9–15.

16.Fudio S, Carcas A, Piñana E, Ortega R: Epileptic sei- zures caused by low valproic acid levels from an interac- tion with meropenem. J Clin Pharm Ther, 2006, 31, 393–396.

17.Glavy JS, Horwitz SB, Orr GA: Identification of the in vivo phosphorylation sites for acidic-directed kinases in murine MDR1b P-glycoprotein. J Biol Chem, 1997, 272, 5909–5914.

18.Goto M, Masuda S, Saito H, Uemoto S, Kiuchi T, Tanaka K, Inui K: C3435T polymorphism in the MDR1 gene affects the enterocyte expression level of CYP3A4 rather than P-gp in recipients of living-donor liver trans- plantation. Pharmacogenetics, 2002, 12, 451–457.

19.Gottesman MM, Hrycyna CA, Schoenlein PV, Germann UA, Pastan I: Genetic analysis of the multidrug trans- porters. Annu Rev Genet, 1995, 29, 607–649.

20.Gottesman MM, Ling V: The molecular basis of mul- tidrug resistance in cancer: the early years of P-glyco- protein research. FEBS Lett, 2006, 580, 998–1009.

21.Haerian BS, Roslan H, Raymond AA, Tan CT, Lim KS, Zulkifli SZ, Mohamed EHM et al.: ABCB1 C3435T polymorphism and the risk of resistance to antiepileptic drugs in epilepsy: a systematic review and meta-analysis.

Seizure, 2010, 19, 339–346.

22.Hodges LM, Markova SM, Chinn LW, Gow JM, Kroetz DL, Klein TE, Altman RB: Very important pharmaco- gene summary: ABCB1 (MDR1, P-glycoprotein). Phar- macogenet Genomics, 2011, 21, 152–161.

23.Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brock- möller J, Johne A, Cascorbi I et al.: Functional polymor- phisms of the human multidrug resistance gene: multiple sequence variations and correlation of one allele with P- glycoprotein expression and activity in vivo. Proc Natl Acad Sci USA, 2000, 97, 3473–3478.

24.Höcht C, Lazarowski A, Gonzales NN, Auzmendi J, Opezzo JA, Bramuglia GF, Taira CA, Girardi E.: Nimo- dipine restores the altered hippocampal phenytoin phar- macokinetics in a refractory epileptic model. Neurosci Lett, 2007, 413, 168–172.

25.Juliano RL, Ling V: A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants.

Biochim Biophys Acta, 1976, 455, 152–162.

26.Kerb R, Aynacioglu AS, Brockmöller J, Schlagenhaufer R, Bauer S, Szekeres T, Hamwi A et al.: The predictive value of MDR1, CYP2C9 and CYPC19 polymorphisms for phenytoin plasma levels. Pharmacogenomics J, 2001, 1, 204–210.

27.Kim DW, Lee SK, Chu K, Jang IJ, Yu KS, Cho JY, Kim SJ: Lack of association between ABCB1, ABCG2, and ABCC2 genetic polymorphisms and multidrug resis- tance in partial epilepsy. Epilepsy Res, 2009, 84, 86–90.

28.Kock K, Grube M, Jedlitschky G, Oevermann L, Sieg- mund W, Ritter CA, Kroemer HK: Expression of adeno- sine triphosphate-binding cassette (ABC) drug transport- ers in peripheral blood cells: relevance for physiology and pharmacotherapy. Clin Pharmacokinet, 2007, 46, 449–470.

29.Kooij G, Mizee MR, van Horssen J, Reijerkerk A, Witte ME, Drexhage JA, van der Pol SM et al.: Adenosine triphosphate-binding cassette transporters mediate che- mokine (C-C motif) ligand 2 secretion from reactive as- trocytes: relevance to multiple sclerosis pathogenesis.

Brain, 2011, 134, 555–570.

30.Kovács R, Raue C, Gabriel S, Heinemann U: Functional test of multidrug transporter activity in hippocampal- neocortical brain slices from epileptic patients. J Neuro- sci Methods, 2011, 200, 164–172.

31.Kwan P, Brodie MJ: Potential role of drug transporters in the pathogenesis of medically intractable epilepsy.

Epilepsia, 2005, 46, 224–235.

32.Kwan P, Schachter SC, Brodie MJ: Drug-resistant epi- lepsy. N Eng J Med, 2011, 365, 919–926.

33.Lasoñ W, Dudra-Jastrzêbska M, Rejdak K, Czuczwar SJ:

Basic mechanisms of antiepileptic drugs and their phar- macokinetic/pharmacodynamic interactions: an update.

Pharmacol Rep, 2011, 63, 271–292.

The multidrug transporter P-glycoprotein

Karolina M. Stêpieñ et al.

(8)

35.Lazarowski A, Czornyj L, Lubieneki F, Gorardi E, Vazquez S, D’Giano C: ABC transporters during epi- lepsy and mechanisms underlying resistance in refrac- tory epilepsy. Epilepsia, 2007, 48, 140–149.

36.Liang LP, Ho YS, Patel M: Mitochondrial superoxide production in the kainate-induced hippocampal damage.

Neuroscience, 2000, 101, 563–570.

37.Löscher W, Klotz U, Zimprich F, Schmidt D: The clini- cal impact of pharmacogenetics on the treatment of epi- lepsy. Epilepsia, 2009, 50, 1–23.

38.Löscher W, Langer O: Imaging of P-glycoprotein func- tion and expression to elucidate mechanisms of pharma- coresistance in epilepsy. Curr Top Med Chem, 2010, 10, 1785–1791.

39.Löscher W, Luna-Tartos C, Romermann K, Fedrowitz M: Do ABC transporters cause pharmacoresistance in epilepsy? Problems and approaches in determining which antiepileptic drugs are affected. Curr Pharm Des, 2011, 17, 2808–2828.

40.Löscher W, Potschka H: Drug resistance in brain dis- eases and the role of drug efflux transporters. Nat Rev Neurosci, 2005, 6, 591–602.

41.Löscher W, Potschka H: Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases. Prog Neurobiol, 2005, 76, 22–76.

42.Löscher W, Potschka H: Role of multidrug transporters in pharmacoresistance to antiepileptic drugs. J Pharma- col Exp Ther, 2002, 301, 7–14.

43.Luna-Tartos C, Fedrowitz M, Löscher W: Evaluation of transport of common antiepileptic drugs by human mul- tidrug resistance-associated proteins (MRP1, 1 and 5) that are overexpressed in pharmacoresistant epilepsy.

Neuropharmacology, 2010, 58, 1019–1032.

44.Luna-Tortos C, Fedrowicz M, Löscher W: Several major antiepileptic drugs are substrates for human P-glyco- protein. Neuropharmacology, 2008, 55, 1364–1375.

45.Mancl EE, Gidal BE: The effect of carbapenem antibiot- ics on plasma concentrations of valproic acid. Ann Phar- macother, 2009, 43, 2082–2087.

46.Meng H, Guo G, Ren J, Zhou H, Ge Y, Guo Y: Effects of ABCB1 polymorphisms on plasma carbamazepine con- centrations and pharmacoresistance in Chinese patients with epilepsy. Epilepsy Behav, 2011, 21, 27–30.

47.Miller DS, Bauer B, Hartz AMS: Modulation of P- glycoprotein at the blood-brain barrier; opportunities to improve CNS pharmacotherapy. Pharmacol Rev, 2008, 60, 196–209.

48.Moerman L, Wyffels L, Slaets D, Raedt R, Boon P, De Vos F: Antiepileptic drugs modulate P-glycoproteins in the brain: a mice study with11C-desmethylloperamide.

Epilepsy Res, 2011, 94, 18–25.

49.Mosyagin I, Runge U, Schroeder HW, Dazert E, Vogel- gesang S, Siegmund W, Warzok RW et al.: Association of ABCB1 genetic variants 3435CT and 2677GT to ABCB1 mRNA and protein expression in brain tissue from refractory epilepsy patients. Epilepsia, 2008, 49, 1555–1561.

roscience, 2004, 129, 349–360.

51.Nurmohamed L, Garcia-Bournissen F, Buono RJ, Shan- non MW, Finkelstein Y: Predisposition to epilepsy – does the ABCB1 gene play a role? Epilepsia, 2010, 51, 1882–1885.

52.Ozgon GO, Bebek N, Gul G, Cine N: Association of MDR1 (C3435T) polymorphism and resistance to carba- mazepine in epileptic patients from Turkey. Eur Neurol, 2007, 59, 67–70.

53.Polgar O, Robey RW, Bates SE: ABCG2: structure, func- tion and role in drug response. Expert Opin Drug Metab Toxicol, 2008, 4, 1–15.

54.Potschka H: Modulating P-glycoprotein regulation: fu- ture perspectives for pharmacoresistant epilepsies? Epi- lepsia, 2010, 51, 1333–1347.

55.Remy S, Beck H: Molecular and cellular mechanisms of pharmacoresistance in epilepsy. Brain, 2006, 129, 18–35.

56.Reynolds EH, Elwes RD, Shorvon SD: Why does epi- lepsy become intractable? Lancet, 1983, 322, 952–954.

57.Rogawski MA: Does P-glycoprotein play a role in phar- macoresistance to antiepileptic drugs? Epilepsy Behav, 2002, 3, 493–495.

58.Ronaldson PT, Ashraf T, Bendayan R: Regulation of multidrug resistance protein 1 by tumor necrosis factor a in cultured glial cells: involvement of nuclear factor-kB and c-Jun N-terminal kinase signaling pathways. Mol Pharmacol, 2010, 77, 644–659.

59.Schmidt D, Löscher W: New developments in antiepilep- tic drug resistance: an integrative view. Epilepsy Curr, 2009, 9, 47–52.

60.Seo T, Ishitsu T, Ueda N, Nakada N, Yurube K, Ueda K, Nakagawa K: ABCB1 polymorphisms influence the re- sponse to antiepileptic drugs in Japanese epilepsy pa- tients. Pharmacogenomics, 2006, 7, 551–561.

61.Sills GJ, Mohanraj R, Butler E, McCrindle S, Colllier L, Wilson EA, Brodie MJ: Lack of association between the C3435T polymorphism in the human multidrug resis- tance (MDR1) gene and response to antiepileptic drug treatment. Epilepsia, 2005, 46, 643–647.

62.Silverman JA: Multidrug-resistance transporters. Pharm Biotechnol, 1999, 12, 353–386.

63.Simon C, Stieger B, Kullak-Ublick GA, Fried M, Muel- ler S, Fritschy JM, Wieser HG, Pauli-Magnus C:Intesti- nal expression of cytochrome P450 enzymes and ABC transporters and carbamazepine and phenytoin disposi- tion. Acta Neurol Scand, 2007, 115, 232–242.

64.Sisodiya S: Drug resistance in epilepsy: not futile, but complex? Lancet Neurol, 2003, 2, 331.

65.Sisodiya SM, Hefferman J, Squier MV: Over-expression of P-glycoprotein in malformations of cortical develop- ment. Neuroreport, 1999, 10, 3437–3441.

66.Sisodiya SM, Lin W-R, Harding BN, Squier MV, Thorn M: Drug resistance in epilepsy: human epilepsy. Novartis Found Symp, 2002, 243, 167–174.

67.Sissung TM, Baum CE, Kirkland CT, Gao R, Gardner ER, Figg WD: Pharmacogenetics of membrane trans-

(9)

porters: an update on current approaches. Mol Biotech- nol, 2010, 44, 152–167.

68.Ufer M, Mosyagin I, Muhle H, Jacobsen T, Haenisch S, Häsler R, Faltraco F et al.: Non-response to antiepileptic pharmacotherapy is associated with the ABCC2-24C-T polymorphism in young and adult patients with epilepsy.

Pharmacogenet Genomics, 2009, 19, 353–362.

69.Vaalburg W, Hendrikse NH, Elsinga PH, Bart J, van Waarde A: P-glycoprotein activity and biological re- sponse. Toxicol Appl Pharmacol, 2005, 207, 257–260.

70.van Vliet EA, Redeker S, Aronica E, Edelbroek PM, Gorter JA: Expression of multidrug transporters MRP1, MRP2, and BCRP shortly after status epilepticus, during the latent period, and in chronic epileptic rats. Epilepsia, 2005, 46, 1569–1580.

71.van Vliet EA, Zibell G, Pekcec A, Schlichtiger J, Edel- broek PM, Holtman L, Aronica E et al.: COX-2 inhibi- tion controls P-glycoprotein expression and promotes brain delivery of phenytoin in chronic epileptic rats.

Neuropharmacology, 2010, 58, 404–412.

72.Warren MS, Zerangue N, WoodfordK, Roberts LM, Tate EH, Feng B, Li C et al.: Comparative gene expression profiles of ABC transporters in brain microvessel endo- thelial cells and brain in five species including human.

Pharmacol Res, 2009, 59, 404–413.

73.Weiss J, Kerpen Ch J, Lindenmaier H, Dormann SM, Haefeli W: Interaction of antiepileptic drugs with human P-glycoprotein in vitro. J Pharmacol Exp Ther, 2003, 307, 262–267.

74.Yang HW, Liu HY, Liu X, Zhang DM, Liu YC, Liu XD, Liu GJ et al.: Increased P-glycoprotein function and level after long-term exposure of four antiepileptic drugs to rat brain microvascular endothelial cells in vitro. Neurosci Lett, 2008, 434, 299–303.

75.Zhang C, Kwan P, Zuo Z, Baum L: In vitro concentration dependent transport of phenytoin and phenobarbital, but not ethosuximide, by human P-glycoprotein. Life Sci, 2010, 86, 899–905.

76.Zhang C, Kwan P, Zuo Z, Baum L: The transport of an- tiepileptic drugs by P-glycoprotein. Adv Drug Deliv Rev, 2011, doi:10.1016/j.addr.2011.12.003.

77.Zhang C, Zuo Z, Kwan, P, Baum L: In vitro transport profile of carbamazepine, oxcarbazepine, eslicarbazepine acetate, and their active metabolites by human P-glyco- protein. Epilepsia, 2011, 52, 1894–1904.

78.Zhang Y, Guo X, Lin ET, Benet LZ: Overlapping sub- strate specificities of cytochrome P450 3A and P-glyco- protein for a novel cysteine protease inhibitor.

Drug Metab Dispos, 1998, 26, 360–366.

79.Zhang Z, Wu JY, Hait WN, Yang JM: Regulation of the stability of P-glycoprotein by ubiquitination. Mol Pharmacol, 2004, 66, 395–403.

Received: January 30, 2012; in the revised form: May 27, 2012;

accepted: June 8, 2012.

The multidrug transporter P-glycoprotein

Karolina M. Stêpieñ et al.

Cytaty

Powiązane dokumenty

OBjECTIvEs The aim of the study was to compare cytotoxic‑based assays with a proliferation assay and drug patch tests in patients with maculopapular eruptions induced

The immunohistochemical staining for p-gp (p-glycoprotein), MRP1 (multidrug resistance associated protein 1), BCRP (breast cancer resistance protein) and LRP (lung resistance

In conclusion, the type II isobolographic analysis used in the present study confirmed a strong influence of 1MeTHIQ on the anticonvulsant efficacy of ETS, GBP and CZP in

When seizure-related events are excluded, patients with epilepsy are only at a higher risk of accidents and injuries than in general population.. This means that proper treatment

Potentiation of extra- synaptic GABA receptors likely contributes to the an- ticonvulsant activity of ethanol, including its protec- tive activity against alcohol withdrawal

The aim of this review is to summarize our knowl- edge on the molecular mechanisms of action, activity profile in animal seizure models, pharmacokinetic profiles, drug interactions

Migraine risk was not related to age of epilepsy onset, but was higher in pa- tients with partial and generalized seizures, and was highest in posttraumatic patients with

Approximately 1/3 of physicians are anxious to employ generic drugs in epilepsy treatment, 2/3 fear deteriorated seizure control following a switch, recur- rent seizures or