Hepatocytes—the choice to investigate drug metabolism and toxicity in man: In vitro variability as a reflection of in vivo

https://doi.org/10.1016/j.cbi.2006.10.013Get rights and content

Abstract

The pharmaceutical industry is committed to marketing safer drugs with fewer side effects, predictable pharmacokinetic properties and quantifiable drug–drug interactions. Drug metabolism is a major determinant of drug clearance and interindividual pharmacokinetic differences, and an indirect determinant of the clinical efficacy and toxicity of drugs. Progressive advances in the knowledge of metabolic routes and enzymes responsible for drug biotransformation have contributed to understanding the great metabolic variations existing in human beings. Phenotypic as well genotypic differences in the expression of the enzymes involved in drug metabolism are the main causes of this variability. However, only a minor part of phenotypic variability in man is attributable to gene polymorphisms, thus making the definition of a normal liver complex. At present, the use of human in vitro hepatic models at early preclinical stages means that the process of selecting drug candidates is becoming much more rational. Cultured human hepatocytes are considered to be the closest model to human liver. However, the fact that hepatocytes are located in a microenvironment that differs from that of the cell in the liver raises the question: to what extent does drug metabolism variability observed in vitro actually reflect that of the liver in vivo? By comparing the metabolism of a model compound both in vitro and in vivo in the same individual, a good correlation between the in vitro and in vivo relative abundance of oxidized metabolites and the hydrolysis of the compound was observed. Thus, it is reasonable to consider that the variability observed in human hepatocytes reflects the existing phenotypic heterogeneity of the P450 expression in human liver.

Introduction

Drugs and other xenobiotics usually have a low solubility in aqueous systems and require biotransformation to metabolites that are more hydrophilic and more readily eliminated. Typically, drug metabolism occurs in two phases: Phase I and Phase II. Phase I of biotransformation is the oxidative pathway in which the compound undergoes oxidation to a more polar substance. Two groups of enzymes, cytochrome P-450 (P450)-depending monooxygenases, a large group of membrane-associated heme proteins and flavin monooxygenases, are major role players in the oxidative metabolism of a wide range of structurally diverse xenobiotics and endogenous compounds. This process is followed by the Phase II reactions in which metabolites are further conjugated by hepatocytes with endogenous molecules by glucuronidation, sulfation, methylation, acetylation and mercapture formation, rendering derivatives that are much more soluble, thus facilitating their elimination [1], [2], [3]. Biotransformation reactions generally follow a detoxification process rendering metabolites inactive. Nevertheless, many drug intermediary products generated during metabolism are highly reactive and toxic, causing hepatotoxicity [4], [5], [6].

The human P450 enzymatic system consists of a great number of different enzymes that demonstrate interindividual variation in activity, which are susceptible to induction and inhibition by a number of compounds. This results in several drug interactions and an increased risk of drug-induced liver injury. The liver is the best-equipped body organ to deal with toxins to prevent or minimize damage caused by reactive intermediates. Although many enzymatic and non-enzymatic pathways of bioinactivation are present in the liver, reactive intermediates may escape the detoxification process and initiate radical-chain reactions. The relationship between bioactivation and the occurrence of hepatic injury is not simple. Such reactive species may either directly or indirectly inflict a toxic injury on the cell by acting as a hapten and initiating an immune-mediated reaction [7], [8].

Drug-induced liver injury is the most frequent cause of post-market withdrawal of an approved drug. Most drug-induced hepatic injuries that occur in humans are unpredictable and poorly understood [5]. A major goal for the pharmaceutical industry is to market safer drugs with fewer side effects, predictable pharmacokinetic properties and quantifiable drug–drug interactions. Drug metabolism is the major source of pharmacokinetic variability in human beings. At the root of this variability are the phenotypic as well genotypic differences in the expression of the enzymes involved in drug metabolism. In addition, qualitative and quantitative interspecies differences in the regulation, expression and functional activity of key ADMET (absorption, distribution, metabolism, excretion and toxicity) processes, particularly P450-mediated metabolism, confound the extrapolation from animals to man. Collectively, deficiencies in ADMET properties and drug-drug interactions are the major causes of attrition during drug development [9], [10]. In vitro assays developed for the evaluation of drug-like properties can accelerate the drug development process. However, there is still an increasing need to develop robust, enhanced-throughput in vitro assays which accurately extrapolate metabolic parameters to humans.

Section snippets

Key issues to be addressed at early stages of drug development

From a commercial perspective, it is desirable that poorly performing compounds are removed early in the discovery phase rather than during the more costly drug development phases. Consequently, over the past decade, in vitro-based strategies in lead optimization screening in conjunction with ADMET screening studies have been incorporated earlier in the drug discovery phase [11], [12]. Studies in the late 1990s indicated that poor pharmacokinetics, metabolism, drug–drug interactions and

P450 variability in human liver

Progressive advances in the knowledge of metabolic routes and enzymes responsible for drug biotransformation have contributed to understanding the great metabolic variations existing in human beings. Phenotypic and genotypic differences in the expression of the enzymes involved in drug metabolism are the main causes of this variability. Analyzing the activity levels of major P450s responsible for drug metabolism in microsomes from a human liver bank revealed the existence of considerable

P450 expression in human hepatocytes

Human hepatocytes in culture show active levels of major P450s involved in drug metabolism [105], [106]. Similarly to that observed in human liver microsomes, high preparation-to-preparation differences in P450 activity levels are found in cultured human hepatocytes from different donors [70], [107], [108], [109] (Fig. 7). This variability is observed in individual P450 enzymes at both activity and mRNA levels. A consequence of this variability, markedly higher than that found in other species,

Drug metabolism by cultured human hepatocytes. How far are we from in vivo?

Human hepatocytes are recognized to be the closest model to the human liver [125]. Once isolated, cells are placed in chemically defined culture conditions where they express typical hepatic biochemical functions, among which is the ability to metabolize drugs [19], [70], [125]. This model is presently considered to be a very useful tool for anticipating drug metabolism and drug hepatotoxicity in man [12], [24], [70], [109]. However, as shown above, the fact that cells are kept in an artificial

Acknowledgements

The authors thank the financial support of the ALIVE Foundation, the Fondo de Investigaciones Sanitarias from Instituto de Salud Carlos III of Spain (03/0339), and the European Commission (LSHB-CT-2004-504761 and LSHB-CT-2004 512051).

References (126)

  • A.P. Li et al.

    Preclinical evaluation of drug–drug interaction potential: present status of the application of primary human hepatocytes in the evaluation of cytochrome P450 induction

    Chem. Biol. Interact.

    (1997)
  • S. Krahenbuhl

    Mitochondria: important target for drug toxicity?

    Hepatology

    (2001)
  • H.J. Zimmerman

    Drug-induced liver disease

    Clin. Liver Dis.

    (2000)
  • D.P. Williams

    Toxicophores: investigations in drug safety

    Toxicology

    (2006)
  • A.T. Banks et al.

    Diclofenac-associated hepatotoxicity: analysis of 180 cases reported to the food and drug administration as adverse reactions

    Hepatology

    (1995)
  • U.A. Boelsterli

    Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity

    Toxicol. Appl. Pharmacol.

    (2003)
  • P.J. O’Brien et al.

    Human and animal hepatocytes in vitro with extrapolation in vivo

    Chem. Biol. Interact.

    (2004)
  • H. Olson et al.

    Concordance of the toxicity of pharmaceuticals in humans and in animals

    Regul. Toxicol. Pharmacol.

    (2000)
  • A.G. Wang et al.

    Effects of phenobarbital on metabolism and toxicity of diclofenac sodium in rat hepatocytes in vitro

    Food Chem. Toxicol.

    (2004)
  • A. Kretz-Rommel et al.

    Diclofenac covalent protein binding is dependent on acyl glucuronide formation and is inversely related to P450-mediated acute cell injury in cultured rat hepatocytes

    Toxicol. Appl. Pharmacol.

    (1993)
  • X. Ponsoda et al.

    Molecular mechanism of diclofenac hepatotoxicity: association of cell injury with oxidative metabolism and decrease in ATP levels

    Toxicol. in Vitro

    (1995)
  • Y. Masubuchi et al.

    Role of motochondrial permeability transition in diclofenac-induced hepatocyte injury in rats

    Hepatology

    (2002)
  • M.S. Miller et al.

    Genetic polymorphisms in human drug metabolic enzymes

    Fundam. Appl. Toxicol.

    (1997)
  • M. Ingelman-Sundberg

    Pharmacogenietics of cytochrome P450 and its applications in drug therapy: the past, present and future

    Trends Pharmacol. Sci.

    (2004)
  • D. Werck-Reichhart et al.

    Cytochromes P450: a success story

    Gen. Biol.

    (2001)
  • P.R. Ortiz de Montellano et al.

    Oxidizing species in the mechanism of cytochrome P450

    Nat. Prod. Rep.

    (2002)
  • D.P. Williams et al.

    Are chemically reactive metabolites responsible for adverse reactions to drugs?

    Curr. Drug Metab.

    (2002)
  • V.J. Navarro et al.

    Drug-related hepatotoxicity

    N. Engl. J. Med.

    (2006)
  • W.M. Lee

    Drug-induced hepatotoxicity

    N. Engl. J. Med.

    (2003)
  • J.L. Walgren et al.

    Role of metabolism in drug-induced idiosyncratic hepatotoxicity

    Crit. Rev. Toxicol.

    (2005)
  • G.P. Aithal et al.

    The use of in vitro methods to predict in vivo pharmacokinetics and drug interactions

    Curr. Drug. Metab.

    (2001)
  • R.J. Riley et al.

    Cellular models for ADMET predictions and evaluation of drug-drug interactions

    Curr. Opin. Drug Discov. Devel.

    (2004)
  • J. Lin et al.

    The role of absorption, distribution, metabolism, excretion and toxicity in drug discovery

    Curr. Top. Med. Chem.

    (2003)
  • D.M. Dambach et al.

    New technologies and screening strategies for hepatotoxicity: use of in vitro models

    Toxicol. Pathol.

    (2005)
  • Z. Yan et al.

    Metabolism profiling, and cytochrome P450 inhibition & induction in drug discovery

    Curr. Top. Med. Chem.

    (2001)
  • T.N. Thompson

    Early ADME in support of drug discovery: the role of metabolic stability studies

    Curr. Drug Metab.

    (2000)
  • C.M. Masimirembwa et al.

    Metabolic stability for drug discovery and development: pharmacokinetic and biochemical challenges

    Clin. Pharmacokinet.

    (2003)
  • P. Baranczewski et al.

    Introduction to in vitro estimation of metabolic stability and drug interactions of new chemical entities in drug discovery and development

    Pharmacol. Rep.

    (2006)
  • E. Kantharaj et al.

    Simultaneous measurement of drug metabolic stability and identification of metabolites using ion-trap mass spectrometry

    Rapid Commun. Mass Spectrom.

    (2003)
  • M.J. Gómez-Lechón et al.

    Human hepatocytes in primary cultures: the choice to investigate drug metabolism in man

    Curr. Drug Metab.

    (2004)
  • R. Gebhardt et al.

    New hepatocyte in vitro systems for drug metabolism: metabolic capacity and recommendations for application in basic research and drug development, standard operation procedures

    Drug Metab. Rev.

    (2003)
  • J.F. Levesque et al.

    Discovery, biosynthesis, and structure elucidation of metabolites of a doping agent and a direct analogue, tetrahydrogestrinone and gestrinone, using human hepatocytes

    Anal. Chem.

    (2005)
  • E.F. Brandon et al.

    Structure elucidation of aplidine metabolites formed in vitro by human liver microsomes using triple quadrupole mass spectrometry

    J. Mass Spectrom.

    (2005)
  • K. Ito et al.

    Comparison of the use of liver models for predicting drug clearance using in vitro kinetic data from hepatic microsomes and isolated hepatocytes

    Pharm. Res.

    (2004)
  • S.J. Griffin et al.

    Prediction of in vitro intrinsic clearance from hepatocytes: comparison of suspensions and monolayer cultures

    Drug Metab. Dispos.

    (2005)
  • D. Hallifax et al.

    Prediction of metabolic clearance using cryopreserved human hepatocytes: kinetic characteristics for five benzodiazepines

    Drug Metab. Dispos.

    (2005)
  • J.B. Houston

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

    Biochem. Pharmacol.

    (1984)
  • Y. Naritomi et al.

    Utility of hepatocytes in predicting drug metabolism: comparison of hepatic intrinsic clearance in rats and humans in vivo and in vitro

    Drug Metab. Dispos.

    (2003)
  • R.J. Riley et al.

    A unified model for predicting human hepatic, metabolic clearance from in vitro intrinsic clearance data in hepatocytes and microsomes

    Drug Metab. Dispos.

    (2005)
  • K. Ito et al.

    Prediction of pharmacokinetic alterations caused by drug–drug interactions: metabolic interaction in the liver

    Pharmacol. Rev.

    (1998)
  • Cited by (121)

    • Drug-metabolizing enzymes and inflammatory responses

      2022, Biochemistry of Drug Metabolizing Enzymes: Trends and Challenges
    View all citing articles on Scopus
    View full text