5
Carbamazepine is not a substrate for P-glycoprotein Andrew Owen, Munir Pirmohamed, Justice N. Tettey, Paul Morgan, 1 David Chadwick 2 & B. Kevin Park Department of Pharmacology and Therapeutics, The University of Liverpool, Ashton Street, Liverpool, L69 3GE, U.K, 1 Department of Drug Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ and 2 Department of Neurological Sciences, The University of Liverpool, Walton Centre for Neurology and Neurosurgery, Lower Lane, Liverpool, L9 7AE Aims To determine whether the anticonvulsant carbamazepine (CBZ), a known CYP3A4 substrate, is also a substrate for the multidrug efflux transporter P-glycoprotein (Pgp). Methods The role of Pgp in the transport of CBZ was assessed in three systems: (a) in mdr1a/1b(x/x) and wild-type mice after administration of 2 mg kg x1 and 20 mg kg x1 , which served as a model for brain penetration; (b) in Caco-2 cells, an in vitro model of the intestinal epithelium that is known to express high Pgp levels; and (c) by flow cytometry in lymphocytes using rhodamine 123, a fluorescent substrate for PgP. Results Brain penetration of both doses of CBZ at 1 h and 4 h was comparable in wild-type and mdr1a/1b(x/x) mice. Transport across the Caco-2 cell monolayer was Pgp-independent, and was not affected by the Pgp inhibitor PSC-833. CBZ had no effect on rhodamine 123 efflux from lymphocytes, in contrast to verapamil, which increased fluorescence intensity fivefold. Conclusion CBZ is not a substrate for Pgp. Its efficacy is unlikely to be affected by Pgp over-expression in the brain. Furthermore, the interaction of CBZ with drugs that modulate both CYP3A4 and Pgp function such as verapamil is probably due to inhibition of CYP3A4 and not Pgp. Keywords: carbamazepine, epilepsy, P-glycoprotein Introduction Epilepsy is a common condition affecting 1% of the UK population [1]. Approximately 30% of epileptics have inadequate control of seizures with drugs [2]. The mechanisms underlying such drug resistance are poorly understood. It has been suggested that over-expression of the multidrug transporter P-glycoprotein (Pgp) in the blood–brain barrier may increase drug efflux and limit access to the epileptic focus [3]. Clearly, this would only be important for drugs that are substrates for Pgp. Carbamazepine (CBZ), a widely used anticonvulsant, is regarded as first line therapy for partial seizures [4]. It is also commonly used in combination with other antiepileptic drugs in the treatment of refractory epilepsy [2]. CBZ undergoes extensive metabolism, with the initial oxidative pathways being catalysed by CYP3A4 and CYP2C8 [5]. There is a well-known overlap between substrates for CYP3A4 and Pgp [6], but it is not known whether CBZ is also a substrate for Pgp. It is also interesting to note that CBZ neurotoxicity can be precipitated by con- comitant administration of verapamil, erythromycin or grapefruit juice [7]. Although this has been ascribed to inhibition of CBZ metabolism by CYP3A4, these compounds are also known inhibitors of Pgp [8]. Thus, it is possible that the interaction with CBZ may be due to inhibition not only of CYP3A4, but also of Pgp. In view of these concerns, in this study we have utilized several model systems to investigate whether CBZ is a substrate for Pgp. Methods Materials Minimal Eagle’s Medium (MEM), RPMI-1640, fetal calf serum, trypsin EDTA and Hanks balanced salt solution Correspondence: Dr M. Pirmohamed, Department of Pharmacology and Therapeutics, The University of Liverpool, Ashton Street, Liverpool, L69 3GE, U.K. Tel.: 0151 7945549; Fax: 0151 7945540; E-mail: [email protected] Received 28 June 2000, accepted 6 December 2000. f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 51, 345–349 345

Carbamazepine is not a substrate for P-glycoprotein

Embed Size (px)

Citation preview

Page 1: Carbamazepine is not a substrate for P-glycoprotein

Carbamazepine is not a substrate for P-glycoprotein

Andrew Owen, Munir Pirmohamed, Justice N. Tettey, Paul Morgan,1 David Chadwick2 & B. Kevin Park

Department of Pharmacology and Therapeutics, The University of Liverpool, Ashton Street, Liverpool, L69 3GE, U.K, 1Department of Drug Metabolism,

P®zer Global Research and Development, Sandwich, Kent, CT13 9NJ and 2Department of Neurological Sciences, The University of Liverpool,

Walton Centre for Neurology and Neurosurgery, Lower Lane, Liverpool, L9 7AE

Aims To determine whether the anticonvulsant carbamazepine (CBZ), a known

CYP3A4 substrate, is also a substrate for the multidrug ef¯ux transporter

P-glycoprotein (Pgp).

Methods The role of Pgp in the transport of CBZ was assessed in three systems: (a)

in mdr1a/1b(x/x) and wild-type mice after administration of 2 mg kgx1 and

20 mg kgx1, which served as a model for brain penetration; (b) in Caco-2 cells, an in

vitro model of the intestinal epithelium that is known to express high Pgp levels; and (c)

by ¯ow cytometry in lymphocytes using rhodamine 123, a ¯uorescent substrate for

PgP.

Results Brain penetration of both doses of CBZ at 1 h and 4 h was comparable in

wild-type and mdr1a/1b(x/x) mice. Transport across the Caco-2 cell monolayer was

Pgp-independent, and was not affected by the Pgp inhibitor PSC-833. CBZ had no

effect on rhodamine 123 ef¯ux from lymphocytes, in contrast to verapamil, which

increased ¯uorescence intensity ®vefold.

Conclusion CBZ is not a substrate for Pgp. Its ef®cacy is unlikely to be affected by Pgp

over-expression in the brain. Furthermore, the interaction of CBZ with drugs that

modulate both CYP3A4 and Pgp function such as verapamil is probably due to

inhibition of CYP3A4 and not Pgp.

Keywords: carbamazepine, epilepsy, P-glycoprotein

Introduction

Epilepsy is a common condition affecting 1% of the UK

population [1]. Approximately 30% of epileptics have

inadequate control of seizures with drugs [2]. The

mechanisms underlying such drug resistance are poorly

understood. It has been suggested that over-expression of

the multidrug transporter P-glycoprotein (Pgp) in the

blood±brain barrier may increase drug ef¯ux and limit

access to the epileptic focus [3]. Clearly, this would only be

important for drugs that are substrates for Pgp.

Carbamazepine (CBZ), a widely used anticonvulsant, is

regarded as ®rst line therapy for partial seizures [4]. It is also

commonly used in combination with other antiepileptic

drugs in the treatment of refractory epilepsy [2]. CBZ

undergoes extensive metabolism, with the initial oxidative

pathways being catalysed by CYP3A4 and CYP2C8 [5].

There is a well-known overlap between substrates for

CYP3A4 and Pgp [6], but it is not known whether CBZ

is also a substrate for Pgp. It is also interesting to note

that CBZ neurotoxicity can be precipitated by con-

comitant administration of verapamil, erythromycin or

grapefruit juice [7]. Although this has been ascribed to

inhibition of CBZ metabolism by CYP3A4, these

compounds are also known inhibitors of Pgp [8]. Thus,

it is possible that the interaction with CBZ may be due to

inhibition not only of CYP3A4, but also of Pgp.

In view of these concerns, in this study we have utilized

several model systems to investigate whether CBZ is

a substrate for Pgp.

Methods

Materials

Minimal Eagle's Medium (MEM), RPMI-1640, fetal calf

serum, trypsin EDTA and Hanks balanced salt solution

Correspondence: Dr M. Pirmohamed, Department of Pharmacology and

Therapeutics, The University of Liverpool, Ashton Street, Liverpool, L69

3GE, U.K. Tel.: 0151 7945549; Fax: 0151 7945540; E-mail: [email protected]

Received 28 June 2000, accepted 6 December 2000.

f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 51, 345±349 345

Page 2: Carbamazepine is not a substrate for P-glycoprotein

(HBSS) were purchased from Gibco BRL (Life technol-

ogies, Paisley, UK). Rhodamine 123, imipramine,

verapamil and CBZ were obtained from Sigma (Poole,

Dorset, UK). Ethyl acetate and acetonitrile were

purchased from Fisher Scienti®c (Loughborough, UK).

Dosing of wild-type and transgenic knockout mice

Wild-type (fvb1) and mdr1a/1b(x/x) knockout mice

(Taconic Farms, Germantown, USA) were administered

CBZ (2 mg kgx1 or 20 mg kgx1 in 60% polyethylene

glycol:H2O; i.p.) 1 and 4 h before being sacri®ced (n=4

in each group). The doses used approximate to those used

clinically, at the start of therapy, and in those on

maintenance CBZ therapy. Blood (1.5 ml) was removed

by cardiac puncture prior to removal of brain. The samples

were frozen at x20uC until analysed by h.p.l.c.

Determination of drug transport in Caco-2 cells

Caco-2 cells (1r106), cultured in MEM supplemented

with 20% (v/v) fetal calf serum were placed in 35 mm,

six well-transwell (0.4 mm ®lters) culture plates (Costar,

Bucks, UK) for approximately 2 weeks prior to transport

experiments to allow the cells to adhere and reach

con¯uence. Fresh medium was added to both apical

(1.5 ml) and basolateral (2.5 ml) compartments every

2±3 days until the cells were 100% con¯uent. Cell

monolayer integrity was assessed by measuring the

transepithelial electrical resistance (TEER) using a Milli-

cell-ERS (electrical resistance system). For the transport

experiments, the cell monolayers were equilibriated in

warm (37uC) HBSS, following which the medium from

all apical and basolateral compartments was removed, and

replaced with CBZ (10 mM in HBSS) or HBSS alone.

Transport was also assessed in the presence of the Pgp

inhibitor PSC-833 (100 mM; a gift from Novartis, Basle),

following a 10 min preincubation period. Incubations

were performed for 1 h, after which the apical and

basolateral compartments were sampled for analysis.

Apparent permeability (Papp) values were then calculated

for both apical to basolateral (PappAtoB), and basolateral to

apical (PappBtoA) movement of compound [9]. A PappBtoA

to PappAtoB ratio of greater than 1.0 was indicative of

active ef¯ux transport of compound by an apical transport

protein such as P-gp.

Papp (cm s)=(Concentration in target compartment)/

(Concentration in origin compartment

rmembrane surface area (cm)rtime (s))

The Caco-2 cells used were shown to express high levels

of Pgp mRNA and protein by RT-PCR and Western

blotting, respectively (data not shown). Furthermore, the

same batch of cells had also been shown to have functional

Pgp activity in our laboratories [9].

Development of an h.p.l.c. method for quanti®cation ofcarbamazepine

Owing to lack of availability of radiolabelled CBZ, an

h.p.l.c. method was developed to allow quanti®cation in

the animal and tissue culture studies. All experiments

were carried out on whole brains, which were weighed

following removal, and then homogenized, prior to

extraction with ethyl acetate (3 mlr3). In initial experi-

ments, CBZ (0.8 mg mlx1) was recovered quantitatively

from spiked samples of brain and whole blood following

the addition of an internal standard (imipramine) and

extraction with ethyl acetate (3 ml, three times) with an

ef®ciency of 96.4t5.9% (n=3). A rectilinear relationship

was obtained between detector responses and CBZ

recovered from spiked samples over the concentration

range 0±1.6 mg mlx1 (r2>0.995, n=3). CBZ and imipra-

mine were separated from endogenous material at ambient

temperature on a Lichrospher1 60 RP-select B column

(Merck, Darmstadt, Germany, 125r4 mm, 5 m). The

mobile phase was composed of acetonitrile and ammo-

nium acetate buffer (25 mM, pH 4.0) and was delivered

at 1 ml minx1 with a Kontron-325 gradient pump

(Kontron Instruments, Massachusetts, USA) equipped

with a Kontron-360 autosampler and Kontron-332 u.v.

detector set at 285 nm. A gradient of 40%-80%

acetonitrile over 15 min was used and CBZ and imipra-

mine eluted at 3.5 and 8.5 min, respectively. Chromato-

grams were acquired and analysed using a Kontron PC

integration package. The limits of detection and quanti-

®cation were 30 ng mlx1 and 75 ng mlx1, respectively,

with intra- and interday assay precision of 97.9% and

98.0%, respectively (n=4). The data presented were all

above the limit of quanti®cation. The amount of CBZ

in whole brain was calculated as:

(YrX/A) ng gx1 of brain tissue

where Y=Total volume of reconstituted extract (ml).

X=Concentration of CBZ in reconstituted extract

(ng mlx1).

A=Weight of murine brain (g).

Effect of carbamazepine on ef¯ux of the ¯uorescent dyerhodamine 123

Lymphocytes were isolated from freshly drawn venous

blood as described previously [10]. Rhodamine 123 ef¯ux

studies were carried out by the method of Pro®t et al. [9].

Brie¯y, lymphocytes were loaded with rhodamine 123

(1.5 mg mlx1; 25min; 4uC) before being washed twice

with RPMI-1640 (1 ml; 1uC). The cells were then

A. Owen et al.

346 f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 51, 345±349

Page 3: Carbamazepine is not a substrate for P-glycoprotein

incubated at 37uC or 4uC for 3 h in 1 ml of dye-free

media to allow dye ef¯ux, while parallel experiments

were performed at 37uC in the presence of either the

positive control, verapamil (30 mM), or CBZ (10 and

100 mM). The cells were then harvested, washed twice in

RPMI-1640, and ®xed before ¯ow cytometric analysis on

an EPICS-XL ¯ow cytometer (Beckman Coulter, Bucks,

UK). The lymphocyte population was electronically gated

to exclude debris. At least 5000 events were collected for

each sample, with cellular rhodamine 123 ¯uorescence

being plotted against the number of events. Data

acquisition was performed using the computer program

EXPO analysis software to determine median FL1

¯uorescence values.

Statistical analysis

All results are presented as meants.d. with 95%

con®dence intervals for the difference between the

means, where appropriate. Statistical analysis was per-

formed by using the unpaired t-test, after con®rming that

the data were normally distributed. A two-tailed P value of

<0.05 was accepted as being signi®cant.

Results

Studies in wild-type and mdr1a/1b(x/x) mice showed

that there were no differences in the brain concentrations

of CBZ after administration of low dose (2 mg kgx1) at

1 h (95% CI for the difference x657±236), and high dose

(20 mg kgx1) at both 1 (95% CI x6104±5004) and 4 h

(95% CI x357±282) (Figure 1).

Within whole blood, CBZ concentrations in wild-type

and knockout mice again did not show any differ-

ence and were as follows: 0.14t0.08 mg mlx1 and

0.40t0.29 mg mlx1 (95% CI for the difference

x0.1±0.6) 1 h after administration of 2 mg kgx1 CBZ

in wild-type and knockout mice, respectively; and after

administration of 20 mg kgx1 CBZ, the concentra-

tions were 7.61t1.35 mg mlx1 and 7.78t3.05 mg mlx1

(95% CIx3.9±4.3) at 1 h, and 0.56t0.18 mg mlx1

and 0.55t0.16 mg mlx1 (95% CI x0.3±0.3) at 4 h in

wild-type and knockout mice, respectively.

After low dose CBZ administration, the whole blood

and brain concentrations at 4 h were below the limit of

sensitivity of the assay.

For studies with Caco-2 cells, all monolayers had

a TEER greater than 200 V cmx2, indicating con¯uence

of the monolayer. The ratio of PappBtoA to PappAtoB

was 0.78t0.18 (n=4 experiments in triplicate), indicat-

ing that CBZ was not actively transported from the

basolateral to apical compartments. This was con®rmed

by the use of the Pgp inhibitor PSC-833, which failed to

alter the ratio (0.87t0.32; 95% CI for the difference

x0.5±0.4).

The effect of CBZ on rhodamine 123 ef¯ux in

lymphocytes was determined by ¯ow cytometry, and

was compared with the effect of verapamil (30 mM)

(Figure 2). In the presence of verapamil, the median

¯uorescence increased ®vefold (P<0.005). In contrast,

CBZ had no effect on ¯uorescence intensity at either

10 mM or 100 mM (Figure 2).

1h

Dose (mg kg–1)WT2

KO2

CB

Z c

once

ntra

tion

(ng

g–1 o

f bra

in)

14000

12000

10000

8000

6000

4000

2000

0

Time 1h 1h 1h 4h 4h

WT20

KO20

WT20

KO20

Figure 1 Brain concentrations of carbamazepine (CBZ) in

wild-type (WT) and mdr1a/1b(x/x) knockout (KO) mice at

1 and 4 h after administration of either low (2 mg kgx1) or high

(20 mg kgx1) doses of CBZ. Results represent mean t s.d. of

determinations in four mice within each group. No signi®cant

differences were found between WT and KO mice at the same

doses and times (Student's t-test).

Control Verapamil(30 µM)

Rat

io F

L1 r

elat

ive

to c

ontr

ol

16

14

12

10

8

6

4

0

2

CBZ(10 µM)

CBZ(100 µM)

*

Figure 2 Ratios of median ¯uorescence intensity (FL1) of

human lymphocytes in the presence of either verapamil or

carbamazepine (CBZ) relative to control lymphocytes. Data are

expressed as mean t s.d. of four separate experiments (performed

in duplicate). Statistical analysis performed by t-test: *P<0.005

(when compared with control).

Short report

f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 51, 345±349 347

Page 4: Carbamazepine is not a substrate for P-glycoprotein

Discussion

P-glycoprotein (Pgp), an ATP-dependent membrane-

bound drug ef¯ux pump that is ubiquitously distributed,

transports a large number of therapeutically and structu-

rally disparate drugs [11]. It ®rst came to prominence as

a mechanism for conferring resistance to cancer che-

motherapy [12]. More recently, it has been suggested that

Pgp may also confer drug resistance in other diseases,

including HIV [13] and epilepsy [3].

Approximately 30% of epileptics become refractory

to drug treatment [2]. The ®nding of high levels of

Pgp expression in surgically resected temporal lobe speci-

mens has led to speculation of its role in the pathogenesis

of refractory epilepsy [3]. It is therefore important to

determine which of the anticonvulsants are Pgp substrates,

as such knowledge would be of obvious therapeutic value.

In this study, we have therefore investigated whether CBZ

is a Pgp substrate using three different model systems that

serve as surrogates for Pgp expression in different tissues.

Transgenic mdr1a/1b(x/x) mice are particularly

bene®cial when looking at brain penetration of drugs.

These knockout mice have provided valuable information

on the role of Pgp in drug disposition in vivo [11]. Our

results showed that there were no differences in brain and

whole blood concentrations of CBZ at both 1 and 4 h

after administration of either high or low doses. This is an

important observation since in patients with refractory

epilepsy where Pgp has been shown to be over-expressed

[3], the use of CBZ should lead to adequate brain

concentrations, and hence no impairment of ef®cacy.

Furthermore, many patients with refractory epilepsy are

already treated with CBZ, and yet are still not adequately

controlled [2]; this by itself indicates that factors other than

(or as well as) Pgp expression are also determinants of

treatment resistance in epilepsy.

The Caco-2 cell monolayer serves as a useful model to

investigate the role of Pgp in determining drug absorption

from the intestine. In accordance with the results obtained

with the mdr1a/1b(x/x) knockout mice, no active

transport of CBZ from the basolateral to apical membranes

was demonstrated indicating that CBZ is not a substrate for

intestinal Pgp. This was further con®rmed by the use

of PSC-833, a potent Pgp inhibitor [14], which failed

to change the directional transport. This observation is of

interest for two main reasons: ®rst, the interaction of CBZ

with verapamil and erythromycin [7] is therefore largely

due to inhibition of CYP3A4, and not Pgp. Taken

together with the fact that CBZ entry into the brain is not

in¯uenced by Pgp, it can be concluded that the interaction

is occurring at the level of the intestinal wall and liver, and

not brain. Second, it provides further evidence that not all

CYP3A4 substrates are also Pgp substrates, and is in

accordance with a recent study, which concluded that the

overlap in substrate speci®cities of CYP3A and Pgp was

nothing more than coincidental [6].

The third method used to assess Pgp-mediated transport

of CBZ was with ¯ow cytometry using the ¯uorescent dye

rhodamine 123, which allowed assessment of function

at the level of the individual cell [9]. This method gives an

indication whether a compound is a Pgp substrate, but

cannot by itself be used to conclusively prove that

a compound is not a substrate since there are multiple

binding sites within the Pgp molecule [15]. Furthermore,

rhodamine 123 ef¯ux may also partly be due to other

transporters such as MRP1 [16]. In accordance with

previous studies, verapamil, a known Pgp inhibitor [8],

interfered with rhodamine 123 ef¯ux from lymphocytes.

However, CBZ had no effect on rhodamine 123 ef¯ux

indicating that it did not compete for transport by Pgp,

or indeed by MRP1.

In conclusion, using three different systems, we have

shown that CBZ is not a substrate for the ef¯ux transporter

Pgp. Thus, its ef®cacy is unlikely to be inadvertently

affected by Pgp over-expression in the blood±brain

barrier. Furthermore, the interaction of verapamil and

other CYP3A4 substrates with CBZ is probably due to

inhibition of CYP3A4 and not Pgp.

AO is in receipt of a PhD studentship from P®zer Global Research

and Development. BKP is a Wellcome Principal Fellow. JNT is

a Glaxo Wellcome Postdoctoral fellow. The authors wish to thank

Novartis for supplying PSC-833.

References

1 Shorvon SD. Epidemiology, classi®cation, natural history, and

genetics of epilepsy. Lancet 1990; 336: 93±96.

2 Kwan P, Brodie MJ. Early identi®cation of refractory epilepsy.

N Engl J Med 2000; 342: 314±319.

3 Tishler DM, Weinberg KI, Hinton DR, Barbaro N, Annett

GM, Raffel C. Mdr1 gene-expression in brain of patients with

medically intractable epilepsy. Epilepsia 1995; 36: 1±6.

4 Chadwick D. Safety and ef®cacy of vigabatrin and

carbamazepine in newly diagnosed epilepsy: a multicentre

randomised double-blind study. Vigabatrin European

Monotherapy Study Group. Lancet 1999; 354: 13±19.

5 Kerr BM, Thummel KE, Wurden CJ, et al. Human liver

carbamazepine metabolism ± role of CYP3A4 and CYP2C8 in

10,11-epoxide formation. Biochem Pharmacol 1994; 47:

1969±1979.

6 Kim RB, Wandel C, Leake B et al. Interrelationship between

substrates and inhibitors of human CYP3A and P-glycoprotein.

Pharm Res 1999; 16: 408±414.

7 Dresser GK, Spence JD, Bailey DG.

Pharmacokinetic-pharmacodynamic consequences and clinical

relevance of cytochrome P450 3A4 inhibition. Clin

Pharmacokinet 2000; 38: 41±57.

8 Wandel C, Kim RB, Kajiji S, Guengerich P, Wilkinson GR,

Wood AJ. P-glycoprotein and cytochrome P-450 3A

inhibition: dissociation of inhibitory potencies. Cancer Res

1999; 59: 3944±3948.

A. Owen et al.

348 f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 51, 345±349

Page 5: Carbamazepine is not a substrate for P-glycoprotein

9 Pro®t L, Eagling VA, Back DJ. Modulation of P-glycoprotein

function in human lymphocytes and Caco-2 cell monolayers

by HIV-1 protease inhibitors. AIDS 1999; 13: 1623±1627.

10 Pirmohamed M, Graham A, Roberts P, et al. Carbamazepine

hypersensitivity: assessment of clinical and in vitro chemical

cross-reactivity with phenytoin and oxcarbazepine. Br J Clin

Pharmacol 1991; 32: 741±749.

11 Schinkel AH, Wagenaar E, Mol CAAM, Vandeemter L.

P-glycoprotein in the blood±brain-barrier of mice in¯uences

the brain penetration and pharmacological activity of many

drugs. J Clin Invest 1996; 97: 2517±2524.

12 Germann UA. P-glycoprotein ± a mediator of

multidrug-resistance in tumor-cells. Eur J Cancer 1996;

32A: 927±944.

13 Kim RB, Fromm MF, Wandel C, et al. The drug

transporter P-glycoprotein limits oral absorption and brain

entry of HIV-1 protease inhibitors. J Clin Invest 1998; 101:

289±294.

14 Atadja P, Watanabe T, Xu H, Cohen D. PSC-833, a frontier in

modulation of P-glycoprotein mediated multidrug resistance.

Cancer Metastasis Rev 1998; 17: 163±168.

15 Buxbaum E. Co-operative binding sites for transported

substrates in the multiple drug resistance transporter Mdr1. Eur

J Biochem 1999; 265: 64±70.

16 Davey MW, Hargrave RM, Davey RA. Comparison of drug

accumulation in P-glycoprotein-expressing and

MRP-expressing human leukaemia cells. Leuk Res 1996;

8: 657±664.

Short report

f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 51, 345±349 349