Elsevier

Toxicology Letters

Volume 138, Issues 1–2, 18 February 2003, Pages 29-49
Toxicology Letters

Utility of physiologically based pharmacokinetic models to drug development and rational drug discovery candidate selection

https://doi.org/10.1016/S0378-4274(02)00374-0Get rights and content

Abstract

The present paper proposes a modeling and simulation strategy for the prediction of pharmacokinetics (PK) of drug candidates by using currently available in silico and in vitro based prediction tools for absorption, distribution, metabolism and excretion (ADME). These methods can be used to estimate specific ADME parameters (such as rate and extent of absorption into portal vein, volume of distribution, metabolic clearance in the liver). They can also be part of a physiologically based pharmacokinetic (PBPK) model to simulate concentration–time profiles in tissues and plasma resulting from the overall PK after intravenous or oral administration. Since the ADME prediction tools are built only on commonly generated in silico and in vitro data, they can be applied already in early drug discovery, prior to any in vivo study. With the suggested methodology, the following advantages of the mechanistic PBPK modeling framework can now be utilized to explore potential clinical candidates already in drug discovery: (i) prediction of plasma (blood) and tissue PK of drug candidates prior to in vivo experiments, (ii) supporting a better mechanistic understanding of PK properties, as well as helping the development of more rationale PK-PD relationships from tissue kinetic data predicted, and hence facilitating a more rational decision during clinical candidate selection, and (iii) the extrapolation across species, routes of administration and dose levels.

Introduction

In drug discovery and development, it is of particular interest to estimate the pharmacokinetic (PK) behavior of drug candidates as early as possible based upon in silico and/or in vitro data to select the most promising compounds for further development. The characterization of a drug's PKs, which is the integral of the time coincident processes of absorption, distribution, metabolism and excretion (ADME), can be elucidated by studying each of the ADME processes individually, or by evaluating the overall PK of a study compound by assembling these processes in one global model. Physiologically based prediction methods have been developed for both evaluation types. The conceptual structure of a generic physiologically based pharmacokinetic (PBPK) model of disposition and absorption is depicted in Fig. 1. PBPK models have been developed and used in the last 30 years for the prediction of PK and PK/PD behavior of drugs and chemicals. Furthermore, the physiologically based nature of the PBPK modeling approach allows us to address mechanistic questions with regard to the PK as well as the concentration-effect relationships. PBPK models are capable of predicting quantitatively disposition in different animal species, including humans. Due to these advantages, PBPK models are routinely applied in chemical risk assessment and, therefore, can be considered as an established methodology in this field (Andersen, 1995, Clewell, 1995). However, the application of PBPK modeling in support of drug discovery and development in the pharmaceutical industry remained very limited, apart from a few examples (for examples see Kawai et al., 1994; Charnick et al., 1995). One of the main reasons for the limited applicability of PBPK models in drug discovery and early development was the need for resources demanding input parameters to characterize ADME, which could not be generated within the business process due to time constraints. Recent developments of in silico/in vitro based prediction tools, in particular for absorption, distribution and hepatic clearance, allowed PBPK modeling to become a real alternative for application already in early stages of drug discovery and development. These early applications of mechanism-based PBPK modeling have the potential to facilitate a more rational decision during the clinical candidate selection process, due to a better mechanistic understanding of the ADME processes. The present work summarizes the current status of ADME prediction tools, with the main focus on absorption, distribution and hepatic metabolic clearance. These prediction tools are combined in a PBPK model to obtain predictions of concentration–time profiles and thereby representing the first attempt to predict oral PKs solely based upon in silico and in vitro data. The oral PBPK model suggested is a further extension of the PBPK model of disposition recently published (Poulin and Theil, 2002b) by adding the absorption input function to the combined models of distribution and liver metabolism (Fig. 1). Finally, a modeling and simulation strategy is presented, using the discussed ADME prediction tools separately, as well as combined in a generic PBPK model for rat and humans, which illustrates the use of the method within the drug discovery and development process. One model compound (propranolol) is shown, where the application of the suggested modeling strategy has been illustrated.

Section snippets

Current status of ADME prediction tools

ADME studies in experimental animals are performed as an important part of the preclinical selection process to identify a lead compound for drug development. Traditionally, these studies are performed as whole animal experiments. The advantage of the in vivo studies is that the ADME behavior is evaluated within an intact organism that allows studying the entire PKs in the same model. However, the main disadvantage of in vivo experiments in animals is that they allow only limited throughput of

Modeling strategy

In Fig. 3, a modeling and simulation strategy is depicted to illustrate how the currently available ADME prediction tools in drug discovery and early development can be used. It consists essentially of two major steps, namely the application of the prediction tools, first separately from each other to predict specific ADME parameters, and then combining their input in a modeling and simulation framework to estimate, in a second step, the overall PKs after oral and intravenous administration.

Illustration of the modeling strategy in Fig. 3 by using propranolol as an example

Generally, in drug discovery, plasma kinetics of potential clinical candidates are obtained for the rat, which can be used to confirm the a priori simulations as a kind of model validation. Here, propranolol has been chosen as an example only to illustrate the application of the ADME prediction tools to estimate, in a first step, the PK parameters of absorption, distribution and hepatic clearance solely based upon in silico and in vitro input data (Table 1). The output information (see Table 2:

References (67)

  • B.A. Ploeger et al.

    Physiologically based modeling of glycyrrhizic acid, a compound subject to presystmic metabolism and enterohepatic cycling

    Toxicol. Appl. Pharmacol.

    (2000)
  • P. Poulin et al.

    A priori prediction of tissue:plasma partition coefficients of drugs to facilitate the use of physiologically based pharmacokinetic model in drug discovery

    J. Pharm. Sci.

    (2000)
  • P. Poulin et al.

    Prediction of pharmacokinetics prior to in vivo studies. I. Mechanism-based prediction of volume of distribution

    J. Pharm. Sci.

    (2002)
  • P. Poulin et al.

    Prediction of pharmacokinetics prior to in vivo studies. II. Generic physiologically based pharmacokinetic model of drug disposition

    J. Pharm. Sci.

    (2002)
  • P. Poulin et al.

    Prediction of adipose tissue plasma partition coefficients for structurally unrelated drugs

    J. Pharm. Sci.

    (2001)
  • R.F. Reinoso et al.

    In vitro studies of teicoplanin binding to rat tissues and erythrocytes

    Eur. J. Pharm. Sci.

    (1998)
  • L.X. Yu et al.

    A compartmental absorption and transit model for estimating oral drug absorption

    Int. J. Pharm.

    (1999)
  • M.C. Allen et al.

    Rapid determination of oral pharmacokinetics and plasma free fraction using cocktail approaches: methods and application

    Pharm. Res.

    (1998)
  • Arundel, P.H., 1997. A multi-compartmental model generally applicable to physiologically-based pharmacokinetics...
  • S. Balaz et al.

    A model-based dependence of the human tissue/blood partition coefficients of chemicals on lipophilicity and tissue composition

    Quant. Struc. Activity Relat.

    (1999)
  • P. Ballard et al.

    Prediction of in vivo tissue distribution from in vitro data. 1. Experiments with markers of aqueous spaces

    Pharm. Res.

    (2000)
  • M. Béliveau et al.

    Concentration dependency of rat blood:air partition coefficients of some volatile organic chemicals

    J. Toxicol. Environ. Health

    (2000)
  • F.M. Belpaire et al.

    Effect of turpentine-induced inflammation on the disposition kinetics of propanolol, metaprolol, and antipyrine in the rat

    Fundam. Clin. Pharmacol.

    (1989)
  • G.E. Blakey et al.

    Quantitative structure-pharmacokinetic relationships. I. Development of a whole-body physiologically based model to characterize changes in pharmacokinetics across a homologous series of barbiturates in the rat

    J. Pharmacokinet. Biopharm.

    (1997)
  • S.B. Charnik et al.

    Perspectives in pharmacokinetics. Physiologically based pharmacokinetic modeling as a tool for drug development

    J. Pharmacokinet. Biopharm.

    (1995)
  • D. Cong et al.

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

    Drug Metab. Dispos.

    (2000)
  • M.T. Donato et al.

    Cytochrome P450 activities in pure and co-cultured rat hepatocytes. Effects of model inducers

    In Vitro Cell. Dev. Biol. Anim.

    (1994)
  • S. Haddad et al.

    A novel liver PBPK model as tool for in vitro–in vivo extrapolations

    Drug Metab. Rev.

    (2001)
  • Haddad, S., Funk, C., 2002. A physiologically based pharmacokinetic model for the liver as a tool for in vitro–in vivo...
  • J.B. Houston

    Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance

    Biochem. Pharmacol.

    (1994)
  • J.B. Houston et al.

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

    Drug Metab. Rev.

    (1997)
  • J.B. Houston et al.

    In vitro–in vivo scaling of CYP kinetic data not consistent with the classical Michaelis–Menten model

    Drug Metab. Dispos.

    (2000)
  • Y. Igari et al.

    Prediction of diazepam disposition in the rat and man by a physiologically based pharmacokinetic model

    J. Pharmacokinet. Biopharm.

    (1983)
  • Cited by (192)

    • Computer-aided biopharmaceutical characterization: Gastrointestinal absorption simulation

      2023, Computer-Aided Applications in Pharmaceutical Technology: Delivery Systems, Dosage Forms, and Pharmaceutical Unit Operations
    • Physiologically-based pharmacokinetic modeling after drug inhalation

      2021, Inhaled Medicines: Optimizing Development through Integration of In Silico, In Vitro and In Vivo Approaches
    • Pharmacodynamics and pharmacokinetics of the novel synthetic opioid, U-47700, in male rats

      2020, Neuropharmacology
      Citation Excerpt :

      It seems obvious that the present study in rats is not directly comparable to a human patient intoxicated with U-47700. Nevertheless, the disparity between the t1/2 measured in rats versus that reported by Koch et al. may reflect a fundamental difference between rats and humans, whereby the former display much faster clearance of most xenobiotics when compared to the latter (Mahmood, 1998; Theil et al., 2003). The current findings characterized the in vivo pharmacokinetics of U-47700 metabolites for the first time in any species.

    View all citing articles on Scopus
    View full text