Intestinal MDR transport proteins and P-450 enzymes as barriers to oral drug delivery

https://doi.org/10.1016/S0168-3659(99)00034-6Get rights and content

Abstract

Cytochrome P-450 3A4 (CYP3A4), the major phase I drug metabolizing enzyme in humans, and the multidrug efflux pump, MDR or P-glycoprotein (P-gp), are present at high levels in the villus tip enterocytes of the small intestine, the primary site of absorption for orally administered drugs. These proteins are induced or inhibited by many of the same compounds and demonstrate a broad overlap in substrate and inhibitor specificities, suggesting that they act as a concerted barrier to drug absorption. A series of studies from our laboratory of cyclosporine and tacrolimus in humans and a novel cysteine protease inhibitor in rats, dosed concomitantly with inhibitors and inducers of CYP3A4 and P-gp, suggest that gut extraction can be modeled using measures of intestinal metabolism and absorption rate, the latter reflecting changes in P-gp. Results evaluating a preliminary model applied to the CYP3A substrate drugs midazolam, indinavir, saquinavir, and rifabutin suggest that the model may be useful for predicting in vivo intestinal metabolism from in vitro data.

Introduction

Intestinal phase I metabolism and active extrusion of absorbed drug have recently been recognized as major determinants of oral drug bioavailability (e.g., Ref. [1]). Many factors are involved in oral drug delivery, yet, the measured oral bioavailability of a particular drug can be broken down into components that reflect delivery to the intestine (gastric emptying, pH, food), absorption from the lumen (dissolution, lipophilicity, particle size, active uptake), intestinal metabolism (phase I and/or phase II enzymes), active extrusion (drug efflux pumps) and finally first-pass hepatic extraction. Major advances have been implemented by pharmaceutical scientists to address the delivery and absorption issues as exemplified by the majority of the excellent papers presented at the June, 1998 symposium honoring Professor Tsuneji Nagai and published in this issue. The importance of the hepatic first pass metabolism and the ability to quantitate the hepatic extraction have been recognized since the mid-1970s [2]. Here, we will concentrate on intestinal phase I metabolism and intestinal active drug efflux.

Both cytochrome P-450 3A4 (CYP3A4), the major phase I drug metabolizing enzyme in humans, and the multidrug efflux pump, MDR or P-glycoprotein (P-gp), are present at high levels in the villus tip enterocytes of the small intestine, the primary site of absorption for orally administered drugs. These proteins are induced or inhibited by many of the same compounds and demonstrate a broad overlap in substrate and inhibitor specificities, suggesting that they act as a concerted barrier to drug absorption [3]. Clinical studies from our laboratory have demonstrated that the bioavailability of three immunosuppressive agents, cyclosporine [4], tacrolimus [5], and sirolimus [in preparation], can be increased by concomitant administration of ketoconazole, a potent CYP3A inhibitor (Ki approximately 1 μM) and an intermediate inhibitor of P-gp (Ki approximately 120 μM).

Conversely, concomitant administration of rifampin, a potent inducer of CYP3A and P-gp, markedly decreased the bioavailability of cyclosporine [6]. A recent clinical study in kidney transplant patients has indicated that variability of intestinal expression of P-gp in humans may be a more important determinant of cyclosporine bioavailability than the variability of intestinal CYP3A [7]. However, the presence of CYP3A is believed to be responsible for the decreased cyclosporine bioavailability. A series of studies in animals have indicated that inhibition of intestinal P-gp has marked effects on the bioavailability of paclitaxel [8], digoxin [9] and HIV-1 protease inhibitors [10], [11]. We have also recently demonstrated the marked increase in bioavailability of an investigational cysteine protease inhibitor when the drug was dosed concomitantly with ketoconazole to rats [12]. Most recently, we have begun to model the effects of CYP3A and P-gp on intestinal absorption and bioavailability, with the final goal of being able to use in vitro measures of drug metabolism by human intestinal CYP3A and bidirectional flux by human MDR transfected cell lines to predict in vivo the extent of gut first-pass extraction.

Section snippets

Intestinal CYP3A4

Enzymes of the CYP3A family are the predominant phase I drug metabolizing species found in humans, accounting for approximately 30% of hepatic CYP [13] and greater than 70% of small intestinal CYP [14]. Moreover, CYP3A is estimated to metabolize more than half of the drugs that are substrates for the P450 system in humans [15], although not always as the only, or even the primary, metabolic enzyme.

The major congener of the CYP3A family is CYP3A4, the predominant form in adult liver and small

Intestinal P-gp

P-gp is the product of the multidrug resistance gene MDR1 in humans and was first characterized as the ATP-dependent transporter responsible for efflux of chemotherapeutic agents from resistant cancer cells [20]. Substrates for P-gp cover a broad range of structures with diverse therapeutic indications. There are no clear structural features defining P-gp substrates, however the molecules tend to be large and amphipathic, containing one or more aromatic rings. P-gp was the first ATP-dependent

Oral drug availability as affected by CYP3A4 and P-gp

The importance of CYP3A and P-gp to oral drug delivery was suggested to us by their joint presence in small intestinal enterocytes as well as by the significant overlap in their substrate specificities [3]. These similarities do not appear to translate into coordinated regulation, however. Lown and coworkers have been unable to find a correlation between intrasubject enterocyte P-gp levels and either enterocyte CYP3A concentration or hepatic CYP3A activity in healthy volunteers [22] or kidney

Recent studies with a novel cysteine protease inhibitor

Morpholine-urea-phenylalanine-homophenylalanine-vinylsulfone-phenyl (K02) is one of a new class of compounds which act as potent cysteine protease inhibitors [24]. Recent in vitro studies in our laboratory [12] have determined that K02 is metabolized by human liver microsomes and recombinantly expressed CYP3A4 with the same metabolite profile. Microsomal metabolism was inhibited by ketoconazole and an antiCYP3A antibody, but not by specific inhibitors of other CYP enzymes [12].

In the same study

Modeling of the effects of intestinal CYP3A4 and P-gp

Oral bioavailability (Foral) is the product of the fraction of the drug dose absorbed (Fa), the fraction of the absorbed dose which passes through the gut into the hepatic portal blood unmetabolized (Fg) and the hepatic first-pass availability (Fh), as given in Eq. 2.Foral=Fa·Fg·Fh

Gut and hepatic availability will be defined as one minus the extraction ratio (ER) at each site.Foral=Fa·(1−ERg)·(1−ERh)

When a drug is dosed intravenuosly, it is possible to calculate the hepatic extraction ratio as

Acknowledgements

Work in Dr. Benet’s laboratory was supported in part by NIH grants GM 26691 and CA 72006.

References (32)

  • L Salphati et al.

    Effects of ketoconazole on digoxin absorption and disposition in rat

    Pharmacology

    (1998)
  • R.B Kim et al.

    The drug transporter P-glycoprotein limits oral absorption and brain entry of HIV-1 protease inhibitors

    J. Clin. Invest.

    (1998)
  • A.E Kim et al.

    Saquinavir, an HIV protease inhibitor, is transported by P-glycoprotein

    J. Pharmacol. Exp. Ther.

    (1998)
  • Y Zhang et al.

    Overlapping substrate specificities of cytochrome P450 3A and P-glycoprotein for a novel cysteine protease inhibitor

    Drug Metab. Dispos.

    (1998)
  • T Shimada et al.

    Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 caucasians

    J. Pharmacol. Exp. Ther.

    (1994)
  • P.B Watkins et al.

    Identification of glucocorticoid-inducible cytochromes P450 in the intestinal mucosa of rats and man

    J. Clin. Invest.

    (1987)
  • Cited by (273)

    • Administration of phytochemical nanoformulations (main routes of drug delivery)

      2022, Phytochemical Nanodelivery Systems as Potential Biopharmaceuticals
    View all citing articles on Scopus
    View full text