The barrier function of CYP3A4 and P-glycoprotein in the small bowel

https://doi.org/10.1016/S0169-409X(97)00041-0Get rights and content

Abstract

CYP3A4 present in small bowel enterocytes can catalyze substantial metabolism of some orally administered drugs and, thus, exerts a first-pass effect. Recent data indicate that the P-glycoprotein (the MDR 1 gene product) in the enterocyte brush border also limits the bioavailability of many of the same drugs that interact with CYP3A. It has been proposed that P-glycoprotein and CYP3A4 may be functionally linked because (a) the two proteins are co-localized within the digestive tract and within enterocytes, (b) they share many of the same substrates and (c) they are co-inducible in response to at least some xenobiotics. There are several potential mechanisms whereby the functions of P-glycoprotein and CYP3A4 could be complimentary. First, Pgp may limit absorption in the proximal small bowel, shifting it to more distal, less catalytically efficient segments that contain lower amounts of CYP3A4. Second, Pgp may function to prolong the duration of absorption. This might increase the duration of exposure of drug to and, hence, the extent of metabolism by enterocyte CYP3A4. Finally, Pgp may preferentially remove from the enterocyte primary drug metabolites that are themselves substrates for CYP3A4. This would limit product inhibition and facilitate primary metabolism catalyzed by CYP3A4. Characterization of the roles of CYP3A4 and Pgp in limiting oral drug availability may be aided by recent success in the development of human intestinal cell lines that stably express both CYP3A4 and Pgp.

Introduction

There is now overwhelming evidence that some orally administered drugs that are substrates for cytochrome P450 3A4 (CYP3A4) can undergo significant first-pass metabolism within small intestinal epithelial cells (enterocytes). These data are reviewed elsewhere in this volume and will not be discussed extensively here. CYP3A4 appears to be the most abundant cytochrome P450 present in human small bowel 1, 2; it is found only in the mature enterocytes lining the villus and is not present in crypt cells (Fig. 1). CYP3A4 present in human enterocytes appears to be functionally and structurally identical to CYP3A4 present in human liver. The complete coding region of intestinal CYP3A4 cDNA has been sequenced in our laboratory and determined to be identical to its liver counterpart (unpublished observations). In addition, the pattern of metabolites produced from several CYP3A4 substrates, and the kinetic properties of the reactions, appear to be essentially identical in human small bowel and liver 3, 4, 5, 6. It therefore appears that catalytic properties of CYP3A4 determined in human liver microsomes, or in recombinant systems, can generally be extrapolated to the small bowel.

The related enzyme, CYP3A5, is also detectable in enterocytes in about 70% of adults, and is not co-regulated with CYP3A4 7, 8, 9, 10. Its structure, function and contribution to first-pass metabolism has not been established. However, CYP3A5 appears to be a minor enterocyte enzyme in most individuals [7].

The potential role of P-glycoprotein (Pgp, the MDR-1 gene product) in determining oral availability of some drugs has only recently been appreciated. Pgp is a versatile xenobiotic pump that was first discovered in cancer cells (reviewed in [11]). Pgp functions to make certain cancer cells resistant to several chemotherapeutic agents by pumping them out of the cells, maintaining intracellular concentrations at sublethal levels (MDR stands for multiple drug resistance). Pgp is expressed in a variety of normal (non-cancerous) human tissues, including liver, brain, adrenal gland, kidney and intestinal tract epithelia [12]. In the small bowel, Pgp is present on the apical membrane of the mature epithelial cells and is not detectable in the crypt cells (Fig. 2). Pgp is oriented in the apical membrane to pump xenobiotics from inside the cells back into the lumen of the intestine (i.e., a “countertransport” function) [13]. In humans (as opposed to rodents), there is only one MDR1 gene, and it is generally assumed that intestinal Pgp is functionally identical to Pgp present in other epithelial cells and in cancer cells. Pgp appears to have approximately the same molecular weight in human intestinal cells as in other cell types (as judged by mobility on polyacrylamide gels) 14, 15, 16. However, the complete coding region of human intestinal Pgp cDNA has not been sequenced to the author's knowledge. Kinetic data regarding chemical inhibition of enterocyte Pgp vs. Pgp in other cell types is sparse and somewhat conflicting 16, 17, 18, 19, 20, 21, 22, 23. It can therefore not be assumed at present that characteristics of Pgp transport determined in cells other than enterocytes will be directly applicable to oral drug absorption.

Section snippets

Effect on drug absorption

The demonstration of Pgp in enterocytes provides a mechanism to account for prior observations concerning intestinal (non-biliary) excretion for certain xenobiotics. For example, erythromycin [24], digoxin [24]and some β-blockers [25]and antibiotics 26, 27, 28, 29undergo active secretion from blood into the small bowel intestinal lumen, and appear to be substrates for Pgp 5, 21, 30. Direct evidence that Pgp inhibits the absorption of orally administered drugs comes from several sources. First,

Effect on cyclosporine bioavailability

The simplest explanation for the observed in vivo findings with cyclosporin A is that Pgp functions to prevent the complete absorption of the drug and that the vast majority of metabolism of cyclosporin occurs in the liver. Although this may be the case, several studies have suggested that the intestine can be a very substantial site for metabolism of orally administered cyclosporin A 6, 39, 40, 41. An alternate possibility is that Pgp may not prevent complete absorption of cyclosporin A, but

Future directions

The potential interactions and synergy between CYP3A4 and P-glycoprotein remain largely speculative, mainly because methods available to study such interactions are limited at present. Mouse knock-out models are likely to provide some insight, but it should be remembered that rodents have two expressed Mdr1 genes and may not be a good model for the human situation. Relatively selective chemical inhibitors of Pgp or CYP3A4 that do not inhibit both protein functions are not generally available.

References (59)

  • V. Carriere et al.

    Expression of cytochrome P-450 3A in ht29-mtx cells and Caco-2 clone tc7

    FEBS Lett.

    (1994)
  • P.B. Watkins et al.

    Identification of glucocorticoid-inducible cytochromes P-450 in the intestinal mucosa of rats and man

    J. Clin. Invest.

    (1987)
  • J.C. Kolars et al.

    Identification of rifampin-inducible P450IIIA4 (CYP3A4) in human small bowel enterocytes

    J. Clin. Invest.

    (1992)
  • M.F. Paine et al.

    First-pass metabolism of midazolam by the human intestine

    Clin. Pharmacol. Ther.

    (1996)
  • F.E. Fitzsimmons et al.

    Selective biotransformation of the HIV protease inhibitor saquinavir by human small intestinal cytochrome P450 3A4

    Drug Metab. Dispos.

    (1997)
  • K.S. Lown et al.

    Interpatient heterogeneity in expression of CYP3A4 and CYP3A5 in small bowel. Lack of prediction by the erythromycin breath test [published erratum appears in Drug Metab. Dispos. (1995) 23(3), following the table of contents].

    Drug Metab. Dispos.

    (1994)
  • S.K. Janardan et al.

    CYP3A5 expression in human enterocytes

    Gastroenterology

    (1994)
  • K.T. Kivisto et al.

    Expression of CYP3A4, CYP3A5 and CYP3A7 in human duodenal tissue

    Br. J. Clin. Pharmacol.

    (1996)
  • J.C. Kolars et al.

    CYP3A gene expression in human gut epithelium

    Pharmacogenetics

    (1994)
  • M.M. Gottesman et al.

    Biochemistry of multidrug resistance mediated by the multidrug transporter

    Ann. Rev. Biochem.

    (1993)
  • F. Thiebaut et al.

    Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues

    Proc. Natl. Acad. Sci. U.S.A.

    (1987)
  • K.S. Lown, R. Mayo, D.S. Blake, L.Z. Benet, P.B. Watkins, Interpatient variation in intestinal Pgp expression...
  • H.C. Lin, J.H. Meyer, Disorders of gastric emptying, in: T. Yamada (Ed.), Textbook of Gastroenterology, Lippincott, New...
  • J. Hunter et al.

    Epithelial secretion of vinblastine by human intestinal adenocarcinoma cell (hct-8 and t84) layers expressing p-glycoprotein

    Br. J. Cancer

    (1991)
  • J. Hunter et al.

    Drug absorption limited by p-glycoprotein-mediated secretory drug transport in human intestinal epithelial Caco-2 cell layers

    Pharm. Res.

    (1993)
  • H. Saitoh et al.

    Possible involvement of multiple P-glycoprotein-mediated efflux systems in the transport of verapamil and other organic cations across rat intestine

    Pharm. Res.

    (1995)
  • P.F. Augustijns et al.

    Evidence for a polarized efflux system in Caco-2 cells capable of modulating cyclosporin A transport

    Biochem. Biophys. Res. Commun.

    (1993)
  • J. Karlsson et al.

    Transport of celiprolol across human intestinal epithelial (Caco-2) cells: mediation of secretion by multiple transporters including p-glycoprotein

    Br. J. Pharm.

    (1993)
  • E.G. Chikhale et al.

    The effect of verapamil on the transport of peptides across the blood–brain barrier in rats: kinetic evidence for an apically polarized efflux mechanism

    J. Pharmacol. Exp. Ther.

    (1995)
  • Cited by (361)

    • Routes of Drug Administration

      2023, Pharmaceutics: Basic Principles and Application to Pharmacy Practice, Second Edition
    • Impact of ageing on the pharmacokinetics and pharmacodynamics of the drugs

      2022, Pharmacokinetics and Toxicokinetic Considerations - Vol II
    View all citing articles on Scopus
    1

    Tel.: 313-936-8080.

    View full text