Predicting the impact of physiological and biochemical processes on oral drug bioavailability

https://doi.org/10.1016/S0169-409X(01)00179-XGet rights and content

Abstract

Recent advances in computational methods applied to the fields of drug delivery and biopharmaceutics will be reviewed with a focus on prediction of the impact of physiological and biochemical factors on simulation of gastrointestinal absorption and bioavailability. Our application of a gastrointestinal simulation for the prediction of oral drug absorption and bioavailability will be described. First, we collected literature data or we estimated biopharmaceutical properties by application of statistical methods to a set of 2D and 3D molecular descriptors. Second, we integrated the differential equations for an advanced compartmental absorption and transit (ACAT) model in order to determine the rate, extent, and approximate gastrointestinal location of drug liberation (for controlled release), dissolution, passive and carrier-mediated absorption, and saturable metabolism and efflux. We predict fraction absorbed, bioavailability, and Cp vs. time profiles for common drugs and compare those estimates to literature data. We illustrate the simulated impact of physiological and biochemical processes on oral drug bioavailability.

Introduction

Application of computational technology during drug discovery and development has the potential to decrease the length of time prior to NDA submission, and reduce the number of experimental procedures required for compound selection and development. Oral bioavailability can be broken down into components that reflect delivery to the intestine (gastric emptying, intestinal transit, local pH, and nutritional status), absorption from the lumen (dissolution, permeability, particle size, intestinal efflux and carrier-mediated transport), first-pass metabolism in the gut, and subsequent first-pass hepatic extraction [1]. Both experimental in vitro and estimated in silico biopharmaceutical properties can be used to predict drug absorption, distribution, metabolism, excretion, and toxicity (ADMET). This review will focus on in silico approaches that have the potential to save valuable resources in drug discovery and development. We review methods that have been developed to build models of individual biopharmaceutical properties, followed by a review of theoretical methods employed to simulate gastrointestinal absorption, hepatic and intestinal metabolism, and pharmacokinetics. Finally, we present our results in predicting bioavailability by estimating biopharmaceutical properties and simulating gastrointestinal absorption and metabolism by extending the advanced compartmental absorption and transit (ACAT) model to account for nonlinear saturable processes.

Section snippets

Computer models of biopharmaceutical properties

In order to lessen the expense and decrease the time associated with experimental determination of in vitro biopharmaceutical properties, existing data has been used to build computational models of octanol–water partition coefficient (log P), effective human jejunal permeability (Peff), cell culture permeability (Caco-2 or MDCK), aqueous solubility, and molecular diffusivity. These models have been developed by application of statistical methods such as linear and partial least squares (PLS)

Simulation of oral drug absorption

Absorption of drugs from the gastrointestinal (GI) tract is very complex and can be influenced by many factors that fall into three classes [2]. The first class represents physicochemical factors including pKa, solubility, stability, diffusivity, lipophilicity, and salt forms. The second class comprises physiological factors including GI pH, gastric emptying, small and large bowel transit times, active transport and efflux, and gut wall metabolism. The third class comprises formulation factors

Metabolism and first pass extraction

The various steps involved in oral drug absorption – dissolution, transit, drug transport and clearance have been traditionally assumed to be first-order non-saturable processes [17]. In reality, some processes are enzyme- or transporter-catalyzed, and they exhibit saturable, nonlinear kinetics. These processes include hepatic and intestinal metabolism, active efflux, and carrier-mediated transport. Apart from strongly influencing drug bioavailability, these nonlinear pharmacokinetic processes,

Bioavailability

Bioavailability is defined as the fraction of administered drug that reaches systemic circulation. Any loss could be due to any of the processes listed above – incomplete absorption, gut metabolism, liver metabolism, etc. If Fi is defined as the fraction of drug that escapes extraction at an organ (or process) i, then the overall bioavailability F is defined asF=i=1nFiIn practice, bioavailability is defined as the ratio of the AUC’s corrected for dose, after extra- and intravascular

Carrier-mediated transport

In order to facilitate absorption of polar nutrients, numerous membrane-bound transport proteins exist in the gastrointestinal tract. Prominent examples include facilitative transporters, the secondary active symporters and antiporters driven by ion gradients, and active ABC (ATP binding cassette) transporters involved in multiple-drug resistance and targeting of antigenic peptides to MHC Class I molecules [61]. Transported substrates range from nutrients and ions to a broad variety of drugs,

Transport, metabolism, and efflux modeling procedures

While many of these processes – gastrointestinal absorption coupled with efflux and transport, liver metabolism, and pharmacokinetics – have been individually modeled, there is still a need to understand all the processes in tandem. Thus, a mathematical model, which incorporates all the nonlinear processes in a systematic manner, will be invaluable in predicting dose–response relationships, and designing optimum dosage regimens for nonlinearly extracted drugs. We have developed a mathematical

Hepatically eliminated drugs (propranolol)

Propranolol is a widely used β-adrenoceptor blocking agent. The conjugate acid form of propranolol is highly soluble in the pH range that exists in the GI tract and it is also rapidly and completely absorbed from the proximal small intestine. Due to high hepatic clearance, the plasma half-life is short – varying from 1.5 to 3 h [73], [74]. After administration of single oral doses, hepatic extraction remains high and much of the dose is eliminated from hepatic portal blood during transfer from

Liver metabolism

Based on literature biopharmaceutical properties and optimized liver Vmax and Km, we predicted the plasma concentration profile, fraction absorbed, and the bioavailability of two different doses of propranolol (IR and ER doses). The predicted quantities agreed well with literature values. Based on steady state plasma concentrations, Wagner et al. [75] estimated a pooled Vmax of approximately 450 mg/day and an apparent Km of 0.05 mg/l [75] while our estimates were 3888 mg/day and 0.05 mg/l (true

Conclusions

  • 1.

    We have developed a new model of saturable processes in oral drug absorption that simulates nonlinear responses in drug bioavailability and pharmacokinetics.

  • 2.

    We have validated the model against experimental data for drugs that undergo liver metabolism alone, gut metabolism and liver metabolism, and efflux and metabolism.

  • 3.

    We used in vitro kinetic constants obtained from homogenate or whole cell experiments under controlled conditions and scaled the constants to the in vivo scenario using

Acknowledgements

We gratefully acknowledge the work of Robert Fraczkiewicz, Grace Fraczkiewicz, and Boyd Steere who helped to collect literature data and developed the application QMPRPlus™ used for estimation of some of the biopharmaceutical properties in this article.

References (106)

  • S.D. Hall et al.

    Molecular and physical mechanisms of first-pass extraction

    Drug. Metab. Dispos.

    (1999)
  • L.X. Yu et al.

    Transport approaches to the biopharmaceutical design of oral drug delivery system: prediction of intestinal absorption

    Adv. Drug Deliv. Rev.

    (1996)
  • M.H. Jacobs

    Some aspects of cell permeability to weak electrolytes

    Cold Spring Harbor Symp. Quant. Biol.

    (1940)
  • C.A.M. Hogben et al.

    On the mechanism of intestinal absorption of drugs

    J. Pharmacol. Exp. Ther.

    (1959)
  • A. Suzuki et al.

    Theoretical model studies of drug absorption and transport in the gastrointestinal tract II

    J. Pharm. Sci.

    (1970)
  • A. Suzuki et al.

    Theoretical model studies of drug absorption and transport in the gastrointestinal tract I

    J. Pharm. Sci.

    (1970)
  • N.F. Ho et al.

    Theoretical model studies of drug absorption and transport in the GI tract (3)

    J. Pharm. Sci.

    (1972)
  • N.F. Ho et al.

    Quantitative interpretation of in vivo buccal absorption of n-alkanoic acids by the physical model approach

    J. Pharm. Sci.

    (1971)
  • J.B. Dressman et al.

    Physicochemical model for dose-dependent drug absorption

    J. Pharm. Sci.

    (1984)
  • A.B. Suttle et al.

    Use of a pharmacokinetic model incorporating discontinuous gastrointestinal absorption to examine the occurrence of double peaks in oral concentration–time profiles

    Pharm. Res.

    (1992)
  • J.D. Wright et al.

    Discontinuous oral absorption pharmacokinetic model and bioavailability of 1-(2-fluoro-5-methyl-beta-l-arabinofuranosyl)uracil (L-FMAU) in rats

    Biopharm. Drug Dispos.

    (1996)
  • D.A. Norris et al.

    Development of predictive pharmacokinetic simulation models for drug discovery

    J. Controlled Release

    (2000)
  • L.X. Yu et al.

    Compartmental transit and dispersion model analysis of small intestinal transit flow in humans

    Int. J. Pharm.

    (1996)
  • L.X. Yu et al.

    A compartmental absorption and transit model for estimating oral drug absorption

    Int. J. Pharm.

    (1999)
  • P.J. Sinko et al.

    Predicting fraction dose absorbed in humans using a macroscopic mass balance approach

    Pharm. Res.

    (1991)
  • D.M. Oh et al.

    Estimating the fraction dose absorbed from suspensions of poorly soluble compounds in humans: a mathematical model

    Pharm. Res.

    (1993)
  • T. Sawamoto et al.

    Prediction of the plasma concentration profiles of orally administered drugs in rats on the basis of gastrointestinal transit kinetics and absorbability

    J. Pharm. Pharmacol.

    (1997)
  • A. Kalampokis et al.

    A heterogeneous tube model of intestinal drug absorption based on probabilistic concepts

    Pharm. Res.

    (1999)
  • A. Kalampokis et al.

    Heterogeneous tube model for the study of small intestinal transit flow

    Pharm. Res.

    (1999)
  • B.R. Stoll et al.

    A theory of molecular absorption from the small intestine

    Chem. Eng. Sci.

    (2000)
  • K. Ito et al.

    Effects of intestinal CYP3A4 and P-glycoprotein on oral drug absorption-theoretical approach

    Pharm. Res.

    (1999)
  • M.F. Paine et al.

    First-pass metabolism of midazolam by the human intestine

    Clin. Pharmacol. Ther.

    (1996)
  • K. Arimori et al.

    Drug exsorption from blood into the gastrointestinal tract

    Pharm. Res.

    (1998)
  • K. Arimori et al.

    The intestinal dialysis of intravenously administered phenytoin by oral activated charcoal in rats

    J. Pharmacobiodyn.

    (1987)
  • W.H. Press

    Numerical recipes in C: the art of scientific computing

    (1992)
  • V. Pade et al.

    Estimation of the relative contribution of the transcellular and paracellular pathway to the transport of passively absorbed drugs in the Caco-2 cell culture model

    Pharm. Res.

    (1997)
  • A.L. Ungell et al.

    Membrane transport of drugs in different regions of the intestinal tract of the rat

    J. Pharm. Sci.

    (1998)
  • A. Adson et al.

    Passive diffusion of weak organic electrolytes across Caco-2 cell monolayers: uncoupling the contributions of hydrodynamic, transcellular, and paracellular barriers

    J. Pharm. Sci.

    (1995)
  • S.M. Pond et al.

    First-pass elimination. Basic concepts and clinical consequences

    Clin. Pharmacokinet.

    (1984)
  • K.T. Le et al.

    First-pass metabolism of lidocaine in the anesthetized rabbit. Contribution of the small intestine

    Drug Metab. Dispos.

    (1996)
  • Y.K. Tam

    Individual variation in first-pass metabolism

    Clin. Pharmacokinet.

    (1993)
  • T. Iwatsubo et al.

    Prediction of in vivo drug metabolism in the human liver from in vitro metabolism data

    Pharmacol. Ther.

    (1997)
  • A.R. Boobis et al.

    Human cytochromes P-450

    Xenobiotica

    (1984)
  • A.R. Boobis et al.

    Multiple forms of human cytochrome P-450

    Biochem. Soc. Trans.

    (1984)
  • T. Lave et al.

    Prediction of hepatic metabolic clearance based on interspecies allometric scaling techniques and in vitro–in vivo correlations

    Clin. Pharmacokinet.

    (1999)
  • T. Lave et al.

    Interspecies scaling of interferon disposition and comparison of allometric scaling with concentration–time transformations

    J. Pharm. Sci.

    (1995)
  • J.B. Houston et al.

    Prediction of hepatic clearance from microsomes, hepatocytes, and liver slices

    Drug Metab. Rev.

    (1997)
  • B.A. Saville et al.

    Models of hepatic drug elimination

    Drug Metab. Rev.

    (1992)
  • B.A. Hoener

    Predicting the hepatic clearance of xenobiotics in humans from in vitro data

    Biopharm. Drug Dispos.

    (1994)
  • T. Lave et al.

    The use of human hepatocytes to select compounds based on their expected hepatic extraction ratios in humans

    Pharm. Res.

    (1997)
  • T. Iwatsubo et al.

    Prediction of in vivo hepatic metabolic clearance of YM796 from in vitro data by use of human liver microsomes and recombinant P-450 isozymes

    J. Pharmacol. Exp. Ther.

    (1997)
  • T. Iwatsubo et al.

    Prediction of in vivo nonlinear first-pass hepatic metabolism of YM796 from in vitro metabolic data

    J. Pharmacol. Exp. Ther.

    (1998)
  • J.J. Bogaards et al.

    Prediction of interindividual variation in drug plasma levels in vivo from individual enzyme kinetic data and physiologically based pharmacokinetic modeling

    Eur. J. Pharm. Sci.

    (2000)
  • G. Schneider et al.

    Combining in vitro and in vivo pharmacokinetic data for prediction of hepatic drug clearance in humans by artificial neural networks and multivariate statistical techniques

    J. Med. Chem.

    (1999)
  • K.S. Lown et al.

    Interpatient heterogeneity in expression of CYP3A4 and CYP3A5 in small bowel. Lack of prediction by the erythromycin breath test

    Drug Metab. Dispos.

    (1994)
  • M.F. Paine et al.

    Characterization of interintestinal and intraintestinal variations in human CYP3A-dependent metabolism

    J. Pharmacol. Exp. Ther.

    (1997)
  • J.C. Kolars et al.

    CYP3A gene expression in human gut epithelium

    Pharmacogenetics

    (1994)
  • Y. Wen et al.

    First-pass disposition of (−)-6-aminocarbovir in rats. I. Prodrug activation may be limited by access to enzyme

    Drug Metab. Dispos.

    (1999)
  • D. Cong et al.

    A new physiologically based, segregated-flow model to explain route-dependent intestinal metabolism

    Drug Metab. Dispos.

    (2000)
  • J. Baron et al.

    Localization, distribution, and induction of xenobiotic-metabolizing enzymes and aryl hydrocarbon hydroxylase activity within lung

    Pharmacol. Ther.

    (1990)
  • Cited by (554)

    • Physiologically based biopharmaceutics modeling of regional and colon absorption in humans

      2023, European Journal of Pharmaceutics and Biopharmaceutics
    • Factors Affecting Drug Absorption and Disposition

      2023, Pharmaceutics: Basic Principles and Application to Pharmacy Practice, Second Edition
    View all citing articles on Scopus
    View full text