Ivonne Importance of P-Glycoprotein - Fund Clin Pharm - 2004

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

doi: 10.1111/j.1472-8206.2004.00291.

REVIEW Functional interaction of intestinal CYP3A4


ARTICLE
and P-glycoprotein
Kari T. Kivistöa*, Mikko Niemia, Martin F. Frommb
a
Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
b
Institute of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Nuremberg,
Germany

Keywords ABSTRACT
absorption,
bioavailability, Intestinal CYP3A4-mediated biotransformation and active efflux of absorbed drug
CYP3A4, by P-glycoprotein are major determinants of bioavailability of orally administered
P-glycoprotein drugs. The hypothesis that CYP3A4 and P-glycoprotein may act in concert to limit
oral drug bioavailability is attractive from a theoretical point of view. Evidence in
support of such an interplay between CYP3A4 and P-glycoprotein comes mainly
Received 6 July 2004;
revised 1 August 2004;
from a limited number of in vitro and animal studies. Obviously, it is a challeng-
accepted 6 September 2004 ing task to demonstrate in vivo in humans that the function of CYP3A4 and
P-glycoprotein in enterocytes is complementary, and results to directly support this
concept remain elusive. However, CYP3A4 and P-glycoprotein are clearly an
*Correspondence and reprints:
integral part of an intestinal defence system to protect the body against harmful
kari.kivisto@ikp-stuttgart.de
xenobiotics, and drugs that are substrates of both proteins often have a low
bioavailability after oral administration. The functional interaction of intestinal
CYP3A4 and P-glycoprotein warrants additional study. Further understanding this
interplay would be potentially useful during drug development to solve bioavail-
ability problems of new drug entities.

extruded from inside the enterocytes into the intestinal


INTRODUCTION
lumen by the P-glycoprotein can be reabsorbed into the
Intestinal oxidative biotransformation and active efflux enterocytes and is thus exposed again to CYP3A4.
of absorbed drug are now recognized as major determi- Furthermore, P-glycoprotein might prevent further
nants of bioavailability of orally administered drugs. interaction of CYP3A4-generated metabolites with the
CYP3A4 is localized in the human small intestine to the enzyme (which might result in product inhibition) by
columnar epithelial cells lining the intestinal lumen [1] extruding them out of the enterocytes [9,10]. Some
and P-glycoprotein is found on the apical surface (that is, examples of drugs that are substrates of both CYP3A4
luminal entrance site) of these cells [2,3]. That CYP3A4 and P-glycoprotein are given in Table I [11,12]. It is
and P-glycoprotein may act synergistically in the small noteworthy that drugs that are substrates of both
intestine to limit oral drug bioavailability is suggested proteins often have a low oral bioavailability. Further
not only by their joint presence at sites of drug understanding the background of the interactive nature
absorption (and elimination) but also by the extensive of intestinal CYP3A4 and P-glycoprotein will therefore
overlap in their substrate specificities [4–7]. The function be important also in controlling and improving oral
of P-glycoprotein may allow CYP3A4 to have repeated bioavailability of CYP3A4/P-glycoprotein substrates [8].
and prolonged access to its substrate molecules [7,8]. The purpose of this review is to briefly summarize the
In other words, a portion of the xenobiotic molecules current knowledge regarding the functional interaction

Ó 2004 Blackwell Publishing Fundamental & Clinical Pharmacology 18 (2004) 621–626 621
622 K. T. Kivistö et al.

Table I Selected substrates of P-glycoprotein. of intestinal CYP3A4 and P-glycoprotein. As it is very


difficult to prove directly in humans that these proteins
P-glycoprotein substrate CYP3A4 substratea
have synergistic actions in limiting the bioavailability of
Anticancer agents orally administered drugs, most studies that have inves-
Docetaxel + tigated this hypothesis have relied on in vitro or animal
Etoposide +
experiments.
Imatinib +
Paclitaxel +
Teniposide +
INTESTINAL CYP3A4
Vinblastine +
Vincristine + CYP3A4, the major isoform of the CYP3A subfamily, is
Beta-blockers one of the most important drug-metabolizing enzymes
Bunitrolol )
in humans. CYP3A4 has a very broad substrate
Carvedilol +
Celiprolol )
specificity and is involved in the phase I metabolism
Talinolol ) of about 50% of currently used drugs. The primary site
Calcium-channel blockers of expression of CYP3A4 is the liver, but intestinal
Diltiazem + enterocytes also contain high amounts of CYP3A4
Mibefradil + [1,13,14]. CYP3A4-mediated biotransformation in the
Verapamil +
gut wall has been shown to substantially contribute to
Other cardiovascular drugs
Digitoxin )
the overall first-pass metabolism of a large number of
Digoxin ) therapeutic drugs [15–21]. For example, a pharmaco-
Quinidine + kinetic study in healthy volunteers showed that the
HIV protease inhibitors first-pass extraction ratios of oral midazolam for the
Amprenavir + gut and liver were comparable (43 and 44%, respect-
Indinavir +
ively, yielding an overall oral bioavailability of about
Nelfinavir +
Saquinavir +
30%) [20]. Similar results were later directly obtained
Ritonavir + for verapamil with a multilumen intestinal perfusion
Corticosteroids catheter in healthy subjects [21]. On the contrary, it
Dexamethasone + has been argued that the role of intestinal drug
Methylprednisolone + metabolism in the first-pass effect may have been
Immunosuppressants
overemphasized [22]. This view was based on problems
Cyclosporin +
Sirolimus +
inherent in the methodology available for determin-
Tacrolimus + ation of in vivo intestinal first-pass metabolism in
Antiemetic drugs humans and the fact that the total CYP3A4 content of
Ondansetron + the human small intestine is small compared with that
Antibiotics of the liver [13]. This apparent discrepancy may partly
Erythromycin +
lie in the anatomical localization of intestinal CYP3A4
Levofloxacin )
Statins
expression [14,22]. Duodenal and proximal jejunal
Atovastatin + mucosa, which are readily exposed to drug molecules
Lovastatin + dissolved in the gastric and intestinal contents, are rich
Antihistamines in villi, the tips of which are lined with mature
Fexofenadine ) CYP3A4-containing enterocytes [1]. This strategic
Terfenadine +
localization of CYP3A4 provides the first site for
Other drugs
metabolism of orally ingested drugs. We recently found
Amitriptyline +
Itraconazole + a much higher CYP3A4 protein content in enterocytes
Losartan ) isolated from human duodenal or jejunal mucosa
Morphine + compared with CYP3A4 content in liver samples
Phenytoin ) obtained from the same subjects [14]. This study thus
Rifampin +
lends further support to the view that metabolism in
a
Indicates whether P-glycoprotein substrates are (+) or are not ()) substrates the gut wall substantially contributes to the overall
of CYP3A4 ([11,12] and references therein). first-pass metabolism of many CYP3A4 substrates.

Ó 2004 Blackwell Publishing Fundamental & Clinical Pharmacology 18 (2004) 621–626


CYP3A4 and P-glycoprotein 623

actions of CYP3A4 and P-glycoprotein during drug


INTESTINAL P-GLYCOPROTEIN
absorption.
P-glycoprotein is a plasma membrane-bound glyco-
protein that belongs to the superfamily of ATP-binding
EVIDENCE OF SYNERGISTIC ACTIONS
cassette (ABC) transporters [23,24]. P-glycoprotein, the
OF CYP3A4 AND P-GLYCOPROTEIN
encoded product of the ABCB1 (MDR1) gene, serves as
IN LIMITING ORAL DRUG
a drug efflux pump that decreases intracellular drug
BIOAVAILABILITY
concentrations. The significance of P-glycoprotein as
a xenobiotic efflux pump was first realized when it The available direct experimental evidence in support of
was found that cancer cells expressing P-glycoprotein the concept that CYP3A4 and P-glycoprotein may act in
are resistant to a large number of anticancer drugs concert to limit oral drug bioavailability originates from
(multidrug resistance, MDR) as a result of active a limited number of in vitro and animal studies.
extrusion of anticancer agents out of the tumour cells Obviously, it is a very difficult if not impossible task to
[25]. P-glycoprotein is also widely expressed in healthy demonstrate in vivo in humans that the function of
tissues, limiting entry (absorption) of drugs and other CYP3A4 and P-glycoprotein in enterocytes is comple-
xenobiotics into the body and specific anatomical mentary. Moreover, factors such as level of expression of
compartments and facilitating their elimination. Drug these proteins along the length of intestine and satura-
absorption was previously considered to be essentially a tion phenomena due to high local drug concentrations
passive process dependent mainly on the physicochem- are likely to modulate the effect of P-glycoprotein on
ical characteristics of the drug (e.g. lipophilicity). intestinal, CYP3A4-mediated drug metabolism in vivo
However, it is now well established that efflux drug [22,27].
transporters such as the P-glycoprotein may reduce Cummins et al. [36] recently set out to determine the
drug absorption [26,27]. P-glycoprotein, which is contribution of P-glycoprotein in determining the extent
localized to the apical membrane of the epithelial cells of CYP3A4-dependent drug metabolism in CYP3A4-
lining the intestinal lumen [2,3], is recognized as a transfected Caco-2 monolayers expressing both CYP3A4
major factor limiting drug bioavailability [27–32]. In and P-glycoprotein, using reasonably selective CYP3A4/
contrast to CYP3A4, the expression of P-glycoprotein P-glycoprotein substrates and inhibitors. It was discov-
appears to increase progressively along the length of ered that when P-glycoprotein alone was inhibited (by
intestine [22,33]. Like CYP3A4, P-glycoprotein exhibits GG918), the extent of metabolism of the dual CYP3A4
a very broad substrate specificity, consistent with a role and P-glycoprotein substrate K77 (a cysteine protease
in transporting compounds absorbed into the entero- inhibitor) was decreased, but there was no change in the
cytes back into the intestinal lumen. extent of metabolism for the exclusive CYP3A4 substrate
felodipine. The authors concluded that P-glycoprotein
can work in concert with CYP3A4 to increase the extent
CO-REGULATION OF CYP3A4 AND
of drug metabolism, by controlling the access of the drug
P-GLYCOPROTEIN
to the intracellular metabolizing enzyme [36]. It should
Several studies have shown that intestinal and hepatic be noted that use of Caco-2 cells to investigate the
CYP3A4 enzymes are not co-ordinately regulated interplay between CYP3A4 and P-glycoprotein may be
[28,34]. This seems to be the case also with P-glyco- problematic because expression of multiple drug efflux
protein [14]. As common regulatory transcription factors transporters and low CYP3A4 content. There are also
such as the orphan nuclear receptor PXR are involved in other studies that have involved in vitro intestinal
the induction of CYP3A4 and P-glycoprotein by, e.g. models and lend support to the interplay hypothesis
rifampicin ([35] and references therein), it has been [9,10,37,38]. In addition, support for this theory has
speculated that the basal intestinal expression of these been obtained from a simulation model [39].
proteins might be co-regulated. However, no significant On the contrary, using CYP3A4-expressing Caco-2
correlation was observed between enterocyte P-glyco- cell monolayers, Mouly et al. [40] recently showed that
protein and CYP3A4 levels in 25 kidney transplant the P-glycoprotein inhibitor LY335979 increased the
recipients [28], suggesting that the expression of CYP3A4 formation rate of a major CYP3A4-mediated metabolite
and P-glycoprotein in the intestine is regulated sepa- (M7) of saquinavir at each saquinavir concentration
rately. This does of course not preclude synergistic tested. The authors suggested that this might have

Ó 2004 Blackwell Publishing Fundamental & Clinical Pharmacology 18 (2004) 621–626


624 K. T. Kivistö et al.

resulted from increased cellular content of saquinavir digoxin metabolism (digoxin is metabolized by CYP3A
due to reduced P-glycoprotein-mediated efflux. However, in the rat) in animals treated with quinidine, a potent
interpretation of this finding is not straightforward as M7 P-glycoprotein inhibitor, compared with the control
is a P-glycoprotein substrate and is subject to secondary group. The authors hypothesized that quinidine
and tertiary metabolism by CYP3A4 ([40] and references increased the extent of digoxin metabolism by blocking
therein). In any event, this study suggests that both the P-glycoprotein-mediated biliary secretion of digoxin
CYP3A4 and P-glycoprotein are involved in the intesti- and thus prolonging exposure of digoxin to hepatic
nal first-pass extraction of saquinavir. CYP3A.
Cummins et al. [41] later extended the findings from
their in vitro study by investigating modulation of
CONCLUSIONS
intestinal CYP3A4-mediated metabolism by P-glyco-
protein in vivo, using an isolated rat single-pass intes- Although the hypothesis that CYP3A4 and P-glyco-
tinal perfusion system with mesenteric cannulation. K77 protein work together to coordinate an absorption
and GG918 were again used as a CYP3A4/P-glycopro- barrier against xenobiotics is attractive from a theoret-
tein substrate and P-glycoprotein inhibitor, respectively, ical point of view (Figure 1), support for this notion
and midazolam was used as an exclusive CYP3A4 largely comes from a small number of in vitro and
substrate. The results further supported a role for animal studies. Direct evidence to support synergism
P-glycoprotein in modulating the extent of intestinal, between these two proteins remains elusive, and the
CYP3A4-mediated biotransformation [41]. quantitative contribution of P-glycoprotein to the intes-
A number of in vitro studies and in vivo rat studies tinal absorption and CYP3A4-mediated metabolism of
performed to characterize the role of intestinal P-glyco-
protein in drug absorption and metabolism have relied
on substrates and inhibitors that affect both CYP3A4
Drug
and P-glycoprotein, precluding differentiation of the
relative contribution of CYP3A4 and P-glycoprotein on Gut lumen Drug metabolite
intestinal drug metabolism.
There is at least one study performed in humans that Apical
indirectly supports the interplay between P-glycoprotein (luminal)
and CYP3A4 in the gut wall [42]. This study, in which ATP P-glycoprotein
the small intestine was perfused with quinidine by means
of a multiluminal perfusion catheter in eight healthy
volunteers, demonstrated the importance of intestinal
P-glycoprotein and CYP3A4 for the plasma concentra-
4
A

tions of quinidine, a substrate of both P-glycoprotein and Enterocyte


P3
Y

CYP3A4. The dose-corrected plasma quinidine AUC from


C

zero to 3 h was negatively correlated with both intestinal


P-glycoprotein content and intestinal CYP3A4 content.
That is, higher amounts of these proteins were associated Basal
with lower plasma quinidine concentrations, indicating
that the oral bioavailability of quinidine was limited by
both P-glycoprotein-mediated efflux and CYP3A4- Figure 1 A scheme of the proposed functional interaction between
mediated metabolism in the gut wall. In addition, as CYP3A4 and P-glycoprotein in human enterocytes. (1) Drug
expected, dose-corrected intraluminal quinidine concen- absorption via passive diffusion or carrier-mediated processes
trations were positively correlated with intestinal from the lumen of the gastrointestinal tract into the enterocyte.
P-glycoprotein expression [42]. (2) CYP3A4-mediated metabolism. (3) Transport of the parent drug
and/or its metabolite(s) from the enterocyte into the gut lumen
It deserves to be mentioned in this context that
by P-glycoprotein. (4) Translocation of the drug and/or its
interplay between CYP3A4 and P-glycoprotein may metabolite(s) across the basal membrane of enterocytes via passive
also occur in the liver, as recently demonstrated by diffusion or carrier-mediated processes into the portal vein.
Lau et al. [43]. They used an isolated perfused rat liver Reproduced from Ref. [26] with the permission of Blackwell
model and found a significant increase in hepatic Publishing (Oxford, UK).

Ó 2004 Blackwell Publishing Fundamental & Clinical Pharmacology 18 (2004) 621–626


CYP3A4 and P-glycoprotein 625

xenobiotics in vivo is not clear. Possible synergism 9 Hochman J.H., Chiba M., Nishime J., Yamazaki M., Lin J.H.
between CYP3A4 and P-glycoprotein does not seem to Influence of P-glycoprotein on the transport and metabo-
lism of indinavir in Caco-2 cells expressing cytochrome
extend to the level of regulation, as there is evidence to
P-450 3A4. J. Pharmacol. Exp. Ther. (2000) 292
suggest that these proteins are not co-ordinately regu-
310–318.
lated in the intestine. The functional interaction of 10 Hochman J.H., Chiba M., Yamazaki M., Tang C., Lin J.H.
intestinal CYP3A4 and P-glycoprotein clearly warrants P-glycoprotein-mediated efflux of indinavir metabolites in
further study. The ensuing knowledge would not only Caco-2 cells expressing cytochrome P450 3A4. J. Pharmacol.
help to better understand the absorption process of orally Exp. Ther. (2001) 298 323–330.
administered drugs but would also be potentially useful 11 Fromm M.F. The influence of MDR1 polymorphisms on
during drug development to solve problems related to P-glycoprotein expression and function in humans. Adv. Drug
Deliv. Rev. (2002) 54 1295–1310.
low bioavailability of new drug entities. Finally, apart
12 Marzolini C., Paus E., Buclin T., Kim R.B. Polymorphisms in
from CYP3A4 and P-glycoprotein, other proteins such as human MDR1 (P-glycoprotein): recent advances and clinical
UDP-glucuronosyltransferases (UGTs) and MDR-associ- relevance. Clin. Pharmacol. Ther. (2004) 75 13–33.
ated protein 2 (MRP2) may also act in concert to 13 Paine M.F., Khalighi M., Fisher J.M. et al. Characterization of
facilitate detoxification of xenobiotics. interintestinal and intraintestinal variations in human CYP3A-
dependent metabolism. J. Pharmacol. Exp. Ther. (1997) 283
1552–1562.
ACKNOWLEDGEMENTS 14 von Richter O., Burk O., Fromm M.F., Thon K.P., Eichelbaum
M., Kivisto K.T. Cytochrome P450 3A4 and P-glycoprotein
The authors’ work was supported by grants from the
expression in human small intestinal enterocytes and hepato-
Robert-Bosch Foundation (Stuttgart) and the Deutsche cytes: a comparative analysis in paired tissue specimens. Clin.
Forschungsgemeinschaft (German Research Foundation, Pharmacol. Ther. (2004) 75 172–183.
Bonn) (FR 1298/2-3). M. Niemi is supported by a 15 Kolars J.C., Awni W.M., Merion R.M., Watkins P.B. First-pass
fellowship from the Alexander von Humboldt Founda- metabolism of cyclosporin by the gut. Lancet (1991) 338
tion (Bonn, Germany). 1488–1490.
16 Hebert M.F., Roberts J.P., Prueksaritanont T., Benet L.Z.
Bioavailability of cyclosporine with concomitant rifampin
REFERENCES administration is markedly less than predicted by hepatic
enzyme induction. Clin. Pharmacol. Ther. (1992) 52
1 Kolars J.C., Lown K.S., Schmiedlin-Ren P. et al. CYP3A gene 453–457.
expression in human gut epithelium. Pharmacogenetics (1994) 17 Wu C.Y., Benet L.Z., Hebert M.F. et al. Differentiation of
4 247–259. absorption and first-pass gut and hepatic metabolism in
2 Thiebaut F., Tsuruo T., Hamada H., Gottesman M.M., Pastan I., humans: studies with cyclosporine. Clin. Pharmacol. Ther.
Willingham M.C. Cellular localization of the multidrug-resist- (1995) 58 492–497.
ance gene product P-glycoprotein in normal human tissues. 18 Fromm M.F., Busse D., Kroemer H.K., Eichelbaum M. Differen-
Proc. Natl. Acad. Sci. USA (1987) 84 7735–7738. tial induction of prehepatic and hepatic metabolism of verap-
3 Greiner B., Eichelbaum M., Fritz P. et al. The role of intestinal amil by rifampin. Hepatology (1996) 24 796–801.
P-glycoprotein in the interaction of digoxin and rifampin. 19 Paine M.F., Shen D.D., Kunze K.L. et al. First-pass metabolism of
J. Clin. Invest. (1999) 104 147–153. midazolam by the human intestine. Clin. Pharmacol. Ther.
4 Wacher V.J., Wu C.Y., Benet L.Z. Overlapping substrate (1996) 60 14–24.
specificities and tissue distribution of cytochrome P450 3A and 20 Thummel K.E., O’Shea D., Paine M.F. et al. Oral first-pass
P-glycoprotein: implications for drug delivery and activity in elimination of midazolam involves both gastrointestinal and
cancer chemotherapy. Mol. Carcinog. (1995) 13 129–134. hepatic CYP3A-mediated metabolism. Clin. Pharmacol. Ther.
5 Benet L.Z., Wu C.Y., Hebert M.F., Wacher V.J. Intestinal drug (1996) 59 491–502.
metabolism and antitransport processes: a potential paradigm 21 von Richter O., Greiner B., Fromm M.F. et al. Determination of
shift in oral drug delivery. J. Control. Release (1996) 39 139– in vivo absorption, metabolism, and transport of drugs by the
143. human intestinal wall and liver with a novel perfusion
6 Watkins P.B. The barrier function of CYP3A4 and P-glycoprotein technique. Clin. Pharmacol. Ther. (2001) 70 217–227.
in the small bowel. Adv. Drug Deliv. Rev. (1997) 27 161–170. 22 Lin J.H., Chiba M., Baillie T.A. Is the role of the small intestine in
7 Benet L.Z., Izumi T., Zhang Y., Silverman J.A., Wacher V.J. first-pass metabolism overemphasized? Pharmacol. Rev. (1999)
Intestinal MDR transport proteins and P-450 enzymes as barriers 51 135–158.
to oral drug delivery. J. Control. Release (1999) 62 25–31. 23 Dietrich C.G., Geier A., Oude Elferink R.P. ABC of oral
8 Zhang Y., Benet L.Z. The gut as a barrier to drug absorption: bioavailability: transporters as gatekeepers in the gut. Gut
combined role of cytochrome P450 3A and P-glycoprotein. (2003) 52 1788–1795.
Clin. Pharmacokinet. (2001) 40 159–168.

Ó 2004 Blackwell Publishing Fundamental & Clinical Pharmacology 18 (2004) 621–626


626 K. T. Kivistö et al.

24 Schwab M., Eichelbaum M., Fromm M.F. Genetic polymorph- bowel. Lack of prediction by the erythromycin breath test. Drug
isms of the human MDR1 drug transporter. Annu. Rev. Metab. Dispos. (1994) 22 947–955.
Pharmacol. Toxicol. (2003) 43 285–307. 35 Fromm M.F. Importance of P-glycoprotein at blood-tissue
25 Gottesman M.M., Fojo T., Bates S.E. Multidrug resistance in barriers. Trends Pharmacol. Sci. (2004) 25 423–429.
cancer: role of ATP-dependent transporters. Nat. Rev. Cancer 36 Cummins C.L., Jacobsen W., Benet L.Z. Unmasking the dynamic
(2002) 2 48–58. interplay between intestinal P-glycoprotein and CYP3A4.
26 Fromm M.F. Importance of P-glycoprotein for drug disposition J. Pharmacol. Exp. Ther. (2002) 300 1036–1045.
in humans. Eur. J. Clin. Invest. (2003) 33(Suppl. 2) 6–9. 37 Gan L.S., Moseley M.A., Khosla B. et al. CYP3A-like cyto-
27 Lin J.H., Yamazaki M. Role of P-glycoprotein in pharmacokinet- chrome P450-mediated metabolism and polarized efflux of
ics: clinical implications. Clin. Pharmacokinet. (2003) 42 59–98. cyclosporin A in Caco-2 cells. Drug Metab. Dispos. (1996) 24
28 Lown K.S., Mayo R.R., Leichtman A.B. et al. Role of intestinal 344–349.
P-glycoprotein (mdr1) in interpatient variation in the oral 38 Johnson B.M., Charman W.N., Porter C.J. The impact of
bioavailability of cyclosporine. Clin. Pharmacol. Ther. (1997) P-glycoprotein efflux on enterocyte residence time and entero-
62 248–260. cyte-based metabolism of verapamil. J. Pharm. Pharmacol.
29 Sparreboom A., van Asperen J., Mayer U. et al. Limited oral (2001) 53 1611–1619.
bioavailability and active epithelial excretion of paclitaxel 39 Ito K., Kusuhara H., Sugiyama Y. Effects of intestinal CYP3A4
(Taxol) caused by P-glycoprotein in the intestine. Proc. Natl. and P-glycoprotein on oral drug absorption: theoretical
Acad. Sci. USA (1997) 94 2031–2035. approach. Pharm. Res. (1999) 16 225–231.
30 Kim R.B., Fromm M.F., Wandel C. et al. The drug transporter 40 Mouly S.J., Paine M.F., Watkins P.B. Contributions of CYP3A4,
P-glycoprotein limits oral absorption and brain entry of HIV-1 P-glycoprotein, and serum protein binding to the intestinal first-
protease inhibitors. J. Clin. Invest. (1998) 101 289–294. pass extraction of saquinavir. J. Pharmacol. Exp. Ther. (2004)
31 Hoffmeyer S., Burk O., von Richter O. et al. Functional 308 941–948.
polymorphisms of the human multidrug-resistance gene: 41 Cummins C.L., Salphati L., Reid M.J., Benet L.Z. In vivo
multiple sequence variations and correlation of one allele with modulation of intestinal CYP3A metabolism by P-glycoprotein:
P-glycoprotein expression and activity in vivo. Proc. Natl. Acad. studies using the rat single-pass intestinal perfusion model.
Sci. USA (2000) 97 3473–3478. J. Pharmacol. Exp. Ther. (2003) 305 306–314.
32 Johne A., Köpke K., Gerloff T. et al. Modulation of steady-state 42 Drescher S., Glaeser H., Murdter T., Hitzl M., Eichelbaum M.,
kinetics of digoxin by haplotypes of the P-glycoprotein MDR1 Fromm M.F. P-glycoprotein-mediated intestinal and biliary
gene. Clin. Pharmacol. Ther. (2002) 72 584–594. digoxin transport in humans. Clin. Pharmacol. Ther. (2003) 73
33 Mouly S., Paine M.F. P-glycoprotein increases from proximal to 223–231.
distal regions of human small intestine. Pharm. Res. (2003) 20 43 Lau Y.Y., Wu C.Y., Okochi H., Benet L.Z. Ex situ inhibition of
1595–1599. hepatic uptake and efflux significantly changes metabolism:
34 Lown K.S., Kolars J.C., Thummel K.E. et al. Interpatient hepatic enzyme-transporter interplay. J. Pharmacol. Exp. Ther.
heterogeneity in expression of CYP3A4 and CYP3A5 in small (2004) 308 1040–1045.

Ó 2004 Blackwell Publishing Fundamental & Clinical Pharmacology 18 (2004) 621–626

You might also like