Elsevier

Toxicology

Volume 216, Issues 2–3, 15 December 2005, Pages 154-167
Toxicology

An in vitro approach to detect metabolite toxicity due to CYP3A4-dependent bioactivation of xenobiotics

https://doi.org/10.1016/j.tox.2005.08.003Get rights and content

Abstract

Many adverse drug reactions are caused by the cytochrome P450 (CYP) dependent activation of drugs into reactive metabolites. In order to reduce attrition due to metabolism-mediated toxicity and to improve safety of drug candidates, we developed two in vitro cell-based assays by combining an activating system (human CYP3A4) with target cells (HepG2 cells): in the first method we incubated microsomes containing cDNA-expressed CYP3A4 together with HepG2 cells; in the second approach HepG2 cells were transiently transfected with CYP3A4. In both assay systems, CYP3A4 catalyzed metabolism was found to be comparable to the high levels reported in hepatocytes. Both assay systems were used to study ten CYP3A4 substrates known for their potential to form metabolites that exhibit higher toxicity than the parent compounds. Several endpoints of toxicity were evaluated, and the measurement of MTT reduction and intracellular ATP levels were selected to assess cell viability. Results demonstrated that both assay systems are capable to metabolize the test compounds leading to increased toxicity, compared to their respective control systems. The co-incubation with the CYP3A4 inhibitor ketoconazole confirmed that the formation of reactive metabolites was CYP3A4 dependent. To further validate the functionality of the two assay systems, they were also used as a “detoxification system” using selected compounds that can be metabolized by CYP3A4 to metabolites less toxic than their parent compounds. These results show that both assay systems can be used to screen for metabolic activation, or de-activation, which may be useful as a rapid and relatively inexpensive in vitro assay for the prediction of CYP3A4 metabolism-mediated toxicity.

Introduction

Adverse reactions associated with exposure of individuals to drugs (ADRs) or xenobiotics are a common and a significant cause of morbidity and mortality (Lazarou et al., 1998, Meyer, 2000). From a clinical perspective, ADRs may be classified as augmented reactions (type A), which are predictable from the known pharmacology and often represent an exaggeration of the pharmacological effects of the drug that may be reversed by dose reduction, or as idiosyncratic reaction (type B), which are unpredictable from the knowledge of the basic pharmacology of the drug and show marked individual susceptibility and no simple dose dependency. From a chemical point of view, this classification can be expanded to reactions (type C), which are predictable from the chemistry of the drug or xenobiotics, and to reactions (type D), which are delayed reactions that occur many years after treatment (Park et al., 1998). Pharmacogenetic, toxicogenetic and other host-dependent factors have been identified to be important in predisposition to type A reactions (Meyer and Gut, 2002). In contrast, most type B, type C and type D ADRs are mediated by toxic metabolites (Gut et al., 1995, Park et al., 1998, Park et al., 2004).

There are many examples of metabolites or reactive intermediates of essentially non-toxic drugs or chemicals that have been shown to exert adverse drug reactions. Some of these ADRs are so serious that drug treatment had to be limited and in some cases the drug had to be withdrawn from the market with enormous consequences for both patients and pharmaceutical industries (e.g. troglitazone, zomepirac and benoxaprofen) (Nassar and Lopez-Anaya, 2004, Wolfgang and Johnson, 2002). Because of this, pharmaceutical companies are making significant efforts to predict ADRs and, currently, several tools and strategies are considered to address the formation of reactive metabolites and the potential consequence for the overall safety profile on candidate drugs.

Reactive metabolites are a common product of phase I oxidation reactions mediated by cytochrome P450 (CYP)-dependent mixed function oxygenases, although also examples of other phase I (e.g. flavin-mono oxygenases; FMOs) and phase II drug metabolizing reactions have been described (Zhou et al., 2005). The generation of such reactive metabolites may produce adverse reactions via different inter-related process such as formation of free radicals, oxidation of thiols and covalent binding with endogenous macromolecules, resulting in the oxidation of cellular components or inhibition of normal cellular functions (Riley et al., 1988). Sometimes, and for unknown reasons, covalently modified proteins may be immunogenic and elicit an immune response (Knowles et al., 2000, Park et al., 2000, Uetrecht, 1999).

In contrast with adverse reactions that are usually dose dependent and due to the pharmacology of the drug (e.g. type A), ADRs caused by toxic metabolites are difficult to predict accurately because metabolic activation of the drug is required to observe the toxic effect. In addition, significant species differences in drug metabolism, chemical instability of the reactive metabolites, and the presence of detoxification pathways are just some of the several factors, which make the screening for toxic metabolites a real challenge.

Over the past years, several approaches have been used to detect metabolism-mediated toxicity in vitro. Because covalent binding of drugs to proteins has been associated with drug toxicity (Zhou et al., 2005), it is rather common practice within large pharmaceutical companies to determine the extent of irreversible binding to protein using radiolabeled drug and human liver microsomes (Day et al., 2005, Kitteringham et al., 1988).

Human hepatocytes represent another in vitro system for the evaluation of metabolism mediated toxicity (Gomez-Lechon et al., 2003, Li et al., 1999), but the poor availability of human liver, the high cost and the significant variability among human hepatocytes preparations make this tool not applicable to an high-throughput screening in drug discovery. More recently, cell lines that have been genetically modified to express a single or multiple drug-metabolising enzyme(s) have been developed (Bull et al., 2001, Dai and Cederbaum, 1995, Lin et al., 1999, Nakagawa et al., 1996, Philip et al., 1999, Wu and Cederbaum, 1996). However, most of these stably transfected cell lines have very low metabolic activity, compared to hepatocytes or liver microsomes. Finally, the immortalized human hepatocyte cell line Fa2N-4 has been recently described as an in vitro tool to study drug metabolism issues (Mills et al., 2004), but like for stably transfected cell lines, also the use of Fa2N-4 cells to address reactive metabolites is limited due to low enzymatic activities.

In the present study, we describe the development of two in vitro models to evaluate the activation of xenobiotics to toxic metabolites. In the first method, we used human CYP3A4 cDNA expressed microsomes (as activating system), in combination with HepG2 cells (as target system).

In the second model, a HepG2 cell line transiently transfected with CYP3A4 was developed. Transient transfection was preferred above stable transfection, because of the higher metabolic activity observed in these cells.

HepG2 cells were selected as target cells for evaluating toxicity because they are derived from human liver and have been extensively used as the test system for the prediction of toxicity, carcinogenicity and cell mutagenicity in humans. Moreover, HepG2 cells contain the co-enzymes NADPH-cytochrome P450 reductase and cytochrome b5, required for CYP mediated drug metabolism (Rodriguez-Antona et al., 2002, Yoshitomi et al., 2001).

Because of the general importance of CYP3A4 in drug metabolism (50% of the drugs in use today are metabolised by CYP3A4) (Guengerich, 2001, Nelson, 1999, Rendic and Di Carlo, 1997) and the several examples of the involvement of CYP3A4 in the formation of reactive metabolites, it was decided to focus on CYP3A4 as an activating system.

In both systems, the metabolic activity of CYP3A4 was determined by incubation with testosterone and midazolam, two well known substrate markers of CYP3A4 metabolism.

For the detection of any effect due to toxic metabolites, reproducible and sensitive endpoints are fundamental. After evaluation of the available assays it was decided that the MTT and ATP were the most suitable endpoints.

The CYP3A4 inhibitor ketoconazole, control microsomes and a control vector plasmid were included to make sure that toxic effects were due to CYP3A4 activity.

To minimize the effect of detoxification, in particular the role of reduced glutathione (GSH), HepG2 cells were treated with l-buthionine S,R-sulphoximine (BSO) in order to deplete GSH levels.

To verify the applicability of both models ten compounds that require metabolic activation to manifest their toxic effects were evaluated. In addition, two compounds known to be metabolised by CYP3A4 into non toxic metabolites were included as well. Compounds tested and their reactive metabolites are reported in Table 1 and compound chemical structures are shown in Fig. 1.

Section snippets

Chemicals

Troglitazone was purchased from Biomol Research Laboratories (Plymouth Meeting, PA, USA); ziprasidone was synthesized in house. All other test compounds were obtained from Sigma (St. Louis, MO, USA).

Cell culture

All cell culture media and supplements were purchased from Invitrogen (Carlsbad, CA, USA). The human hepatic carcinoma cell line HepG2 was obtained from the European Collection of Animal Cell Cultures (ECACC no. 85011430, Porton Down, UK) and cells were used between passage 25 and 28. HepG2 cells

Cell viability

Increasing concentrations of cell lysis buffer (10% Triton® X100), a non-selective cell damaging agent, were used to evaluate the applicability of MTT and ATP as cytotoxicity endpoints. Cell viability, expressed as percentage of non-treated cells (control: 100% viability) in HepG2 cells after 24 h incubation with increasing concentrations of cell lysis buffer is shown in Fig. 2A. For both endpoints a dose-dependent decrease in cell viability was observed.

In order to establish the maximal

Discussion

The biotransformation of xenobiotics to reactive intermediates or toxic metabolites is considered to be a major cause of adverse drug reactions observed in the clinic (Gad, 2003). As a result of this, both regulatory authorities as well as pharmaceutical industries, are currently paying significant attention to address metabolism-mediated toxicity.

Over the past years, several methods have been proposed to predict the potential of chemicals to form toxic metabolites, but until now there is no

Acknowledgements

The authors wish to thank Ms. Anna Moscone for LC–MS analyses, Dr. Roberta Cunati for her support and encouragement and Dr. Italo Poggesi for critical reading of the manuscript.

References (50)

  • C. Beedham et al.

    Ziprasidone metabolism, aldehyde oxidase, and clinical implications

    J. Clin. Psychopharmacol.

    (2003)
  • A. Berson et al.

    Metabolic activation of the nitroaromatic antiandrogen flutamide by rat and human cytochromes P-450, including forms belonging to the 3A and 1A subfamilies

    J. Pharmacol. Exp. Ther.

    (1993)
  • S. Bull et al.

    A Genetically engineered cell-based system for detecting metabolism-mediated toxicity

    Altern. Lab. Anim.

    (2001)
  • M.D. Coleman

    Dapsone toxicity: some current perspectives

    Gen. Pharmacol.

    (1995)
  • Y. Dai et al.

    Cytotoxicity of acetaminophen in human cytochrome P4502E1-transfected HepG2 cells

    J. Pharmacol. Exp. Ther.

    (1995)
  • S.H. Day et al.

    A semi-automated method for measuring the potential for protein covalent binding in drug discovery

    J. Pharmacol. Toxicol. Methods

    (2005)
  • D.C. Evans et al.

    Drug-protein adducts: an industry perspective on minimizing the potential for drug bioactivation in drug discovery and development

    Chem. Res. Toxicol.

    (2004)
  • S.C. Gad

    Active drug metabolites in drug development

    Curr. Opin. Pharmacol.

    (2003)
  • M.J. Gomez-Lechon et al.

    Human hepatocytes as a tool for studying toxicity and drug metabolism

    Curr. Drug Metab.

    (2003)
  • F.P. Guengerich

    Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity

    Chem. Res. Toxicol.

    (2001)
  • J. Gut et al.

    Molecular mimicry in halothane hepatitis: biochemical and structural characterization of lipoylated autoantigens

    Toxicology

    (1995)
  • A. Holownia et al.

    Tamoxifen cytotoxicity in hepatoblastoma cells stably transfected with human CYP3A4

    Biochem. Pharmacol.

    (2004)
  • A. Jaeger et al.

    Clinical features and management of poisoning due to antimalarial drugs

    Med. Toxicol. Adverse Drug Exp.

    (1987)
  • N.R. Kitteringham et al.

    A comparative study of the formation of chemically reactive drug metabolites by human liver microsomes

    Br. J. Clin. Pharmacol.

    (1988)
  • S.R. Knowles et al.

    Idiosyncratic drug reactions: the reactive metabolite syndromes

    Lancet

    (2000)
  • V.E. Kostrubsky et al.

    The use of human hepatocyte cultures to study the induction of cytochrome P-450

    Drug Metab. Dispos.

    (1999)
  • J. Lazarou et al.

    Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies

    JAMA

    (1998)
  • A.P. Li et al.

    Cryopreserved human hepatocytes: characterization of drug-metabolizing enzyme activities and applications in higher throughput screening assays for hepatotoxicity, metabolic stability, and drug-drug interaction potential

    Chem. Biol. Interact.

    (1999)
  • H.L. Lin et al.

    N-Nitrosodimethylamine-mediated cytotoxicity in a cell line expressing P450 2E1: evidence for apoptotic cell death

    Toxicol. Appl. Pharmacol.

    (1999)
  • U.A. Meyer

    Pharmacogenetics and adverse drug reactions

    Lancet

    (2000)
  • U.A. Meyer et al.

    Genomics and the prediction of xenobiotic toxicity

    Toxicology

    (2002)
  • X.S. Miao et al.

    Determination of carbamazepine and its metabolites in aqueous samples using liquid chromatography-electrospray tandem mass spectrometry

    Anal. Chem.

    (2003)
  • J.B. Mills et al.

    Induction of drug metabolism enzymes and MDR1 using a novel human hepatocyte cell line

    J. Pharmacol. Exp. Ther.

    (2004)
  • T. Nakagawa et al.

    Stable expression of human CYP2E1 in Chinese hamster cells: high sensitivity to N,N-dimethylnitrosamine in cytotoxicity testing

    Mutat. Res.

    (1996)
  • A.E. Nassar et al.

    Strategies for dealing with reactive intermediates in drug discovery and development

    Curr. Opin. Drug Discov. Dev.

    (2004)
  • Cited by (0)

    1

    Present address: Drug profiling, Sienabiotech S.p.A., via Fiorentina, 1, 53100 Siena, Italy.

    2

    Present address: Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA 94080, USA.

    View full text