new technologies and screening strategies for ... et al... · discovery toxicology, ... abstract...

10
Toxicologic Pathology, 33:17–26, 2005 Copyright C by the Society of Toxicologic Pathology ISSN: 0192-6233 print / 1533-1601 online DOI: 10.1080/01926230590522284 New Technologies and Screening Strategies for Hepatotoxicity: Use of In Vitro Models DONNA M. DAMBACH,BARBARA A. ANDREWS, AND FREDERIC MOULIN Discovery Toxicology, Bristol-Myers Squibb Pharmaceutical Research Institute, Princeton, New Jersey 08543, USA ABSTRACT Hepatotoxicity remains a significant cause for drug failures during clinical trials. This is due, in part, to the idiosyncratic nature of toxicity in humans and inherent physiological differences between humans and preclinical species leading to limited correct prediction of adverse responses in humans. To address this issue, robust screening assays are being developed, which have heightened predictive capacity for human hepatotoxicity, and may be utilized throughout the discovery and development phases in conjunction with traditional in vivo methods, for decision making during drug selection and risk assessment. This manuscript describes an example application of in vitro-based strategies using human hepatocyte cultures in lead optimization screening in conjunction with ADME profiling, for evaluation of compound-associated CYP450 induction potential, and the identification of potentially useful biomarkers as predictors of hepatotoxicity for use in vitro, and in preclinical species and humans. Keywords. Hepatotoxicity; in vitro cultures; CYP450 induction; biomarker; hepatocytes; screening. INTRODUCTION Drug-induced hepatotoxicity represents a major clinical problem accounting for 50% of all cases of acute liver failure. Although the majority of cases of acute liver failure are due to intentional or unintentional misuse, 16% are idiosyncratic (Bissell et al., 2001; Lee, 2003). At issue is not intrinsic tox- icity, which is well correlated across species (Zimmerman, 1999a; Bissell et al., 2001), but idiosyncratic toxicity. Con- tributing to the difficulty in predicting idiosyncratic events is the rare incidence in humans, 1 in 10,000 to 1 in 100,000, and the diverse mechanisms of toxicity, which are multifacto- rial and related to individual-specific responses (Lee, 2003). Further, the number of animals used in toxicology studies make it difficult to detect hepatotoxicity occurring at a low incidence, as it is impractical to appropriately power such studies. Although a few retrospective studies examining the correlation between the occurrence of hepatotoxicity in hu- mans and in animal studies have been performed, sufficient data are not available to reliably assess the value of preclini- cal animal studies to predict hepatotoxicity in humans (Ballet et al., 1997; Olson et al., 2000). As such, it is understandable that preclinical animal toxicity studies may not be sufficient as the only modeling systems used to predict idiosyncratic toxicity. Address correspondence to: Dr. Donna M. Dambach, Discovery Toxicology, Bristol-Myers Squibb, PRI, P.O. Box 4000, F14-03, Princeton, New Jersey 08543, USA; e-mail: [email protected] Abbreviations: CYP450: cytochrome P450; UDPGT: uridine phosphoglucuronosyltransferase; CMV: cytomegalovirus; IC 50 : 50% inhibitory concentration; MTS: 3-(4,5-dimethylthiazol-2-yl)-5-(3- carboxymethoxyphenyl)-2-(4-sulphophenyl)-H-tetrazolium, inner salt; ATP: adenosine triphosphate; DMSO: dimethyl sulfoxide; µM: micromo- lar; ED 50 : 50% effective dose; SAR: structure activity relationship; ADME: absorption, distribution, metabolism, elimination; TZD: thiazolidinedione; PCA: principal component analysis; LC/LC/MS/MS: multidimensional packed capillary high performance liquid chromotography coupled tandem mass spectrometry; MIF: macrophage migration inhibitory factor; ELISA: enzyme linked immunosorbant assay; VOD: veno-occlusive disease. Therefore, in continuing efforts to improve the identifica- tion of potential hepatotoxicity issues during lead optimiza- tion, the addition of new, predictive assays to the discovery armamentarium is necessary. In vitro systems are an impor- tant component of these new assay system applications. Use of in vitro systems is not new to investigative toxicology or lead optimization processes. Systems such as primary cell cultures, immortalized cell lines, liver slices and whole, per- fused livers are well-established models used for investiga- tive work (Acosta et al., 1985; Butterworth et al., 1989; Ulrich et al., 1995; Guillouzo et al., 1997; Groneberg et al., 2002). Likewise, the use of HepG2 cell lines and primary hepatocyte cell cultures to assess potential cytotoxicity are promoted by several commercial laboratories. It is the development of up- dated screening paradigms, which combine analysis of data generated by new or modified in vitro assays with estab- lished in vivo methods, that may generate additional insights to help guide the risk assessment/decision-making process. In combination, these assays provide a more well-rounded assessment of potential risk. This manuscript will present an example of a multi-tier paradigm using in vitro cultures of immortalized cell lines and primary hepatocyte cultures to assess potential risk for acute clinical liabilities, and demonstrate how the results from these systems are used to help make recommendations regarding lead optimization and the potential for clinical hepatotoxicity (acute liver failure). Further, the use of these in vitro models to assess CYP450 enzyme induction potential and biomarker identification will also be described. In Vitro Cell Systems The cell-based systems described herein include immortal- ized cell lines and primary hepatocyte cultures. The immor- talized cell system is comprised of a series of five cell lines uti- lized together as an assay system. These cells were originally derived from normal human hepatocytes (Pfeifer et al., 1993). The parent cell line has no significant phase I and phase II me- tabolizing capabilities based upon in-house characterization 17

Upload: vantram

Post on 27-Jul-2018

214 views

Category:

Documents


0 download

TRANSCRIPT

Toxicologic Pathology, 33:17–26, 2005Copyright C© by the Society of Toxicologic PathologyISSN: 0192-6233 print / 1533-1601 onlineDOI: 10.1080/01926230590522284

New Technologies and Screening Strategies for Hepatotoxicity:Use of In Vitro Models

DONNA M. DAMBACH, BARBARA A. ANDREWS, AND FREDERIC MOULIN

Discovery Toxicology, Bristol-Myers Squibb Pharmaceutical Research Institute, Princeton, New Jersey 08543, USA

ABSTRACT

Hepatotoxicity remains a significant cause for drug failures during clinical trials. This is due, in part, to the idiosyncratic nature of toxicity inhumans and inherent physiological differences between humans and preclinical species leading to limited correct prediction of adverse responses inhumans. To address this issue, robust screening assays are being developed, which have heightened predictive capacity for human hepatotoxicity,and may be utilized throughout the discovery and development phases in conjunction with traditional in vivo methods, for decision making duringdrug selection and risk assessment. This manuscript describes an example application of in vitro-based strategies using human hepatocyte culturesin lead optimization screening in conjunction with ADME profiling, for evaluation of compound-associated CYP450 induction potential, and theidentification of potentially useful biomarkers as predictors of hepatotoxicity for use in vitro, and in preclinical species and humans.

Keywords. Hepatotoxicity; in vitro cultures; CYP450 induction; biomarker; hepatocytes; screening.

INTRODUCTION

Drug-induced hepatotoxicity represents a major clinicalproblem accounting for 50% of all cases of acute liver failure.Although the majority of cases of acute liver failure are dueto intentional or unintentional misuse, 16% are idiosyncratic(Bissell et al., 2001; Lee, 2003). At issue is not intrinsic tox-icity, which is well correlated across species (Zimmerman,1999a; Bissell et al., 2001), but idiosyncratic toxicity. Con-tributing to the difficulty in predicting idiosyncratic events isthe rare incidence in humans, 1 in 10,000 to 1 in 100,000,and the diverse mechanisms of toxicity, which are multifacto-rial and related to individual-specific responses (Lee, 2003).Further, the number of animals used in toxicology studiesmake it difficult to detect hepatotoxicity occurring at a lowincidence, as it is impractical to appropriately power suchstudies. Although a few retrospective studies examining thecorrelation between the occurrence of hepatotoxicity in hu-mans and in animal studies have been performed, sufficientdata are not available to reliably assess the value of preclini-cal animal studies to predict hepatotoxicity in humans (Balletet al., 1997; Olson et al., 2000). As such, it is understandablethat preclinical animal toxicity studies may not be sufficientas the only modeling systems used to predict idiosyncratictoxicity.

Address correspondence to: Dr. Donna M. Dambach, DiscoveryToxicology, Bristol-Myers Squibb, PRI, P.O. Box 4000, F14-03, Princeton,New Jersey 08543, USA; e-mail: [email protected]

Abbreviations: CYP450: cytochrome P450; UDPGT: uridinephosphoglucuronosyltransferase; CMV: cytomegalovirus; IC50: 50%inhibitory concentration; MTS: 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphophenyl)-H-tetrazolium, inner salt;ATP: adenosine triphosphate; DMSO: dimethyl sulfoxide; µM: micromo-lar; ED50: 50% effective dose; SAR: structure activity relationship; ADME:absorption, distribution, metabolism, elimination; TZD: thiazolidinedione;PCA: principal component analysis; LC/LC/MS/MS: multidimensionalpacked capillary high performance liquid chromotography coupled tandemmass spectrometry; MIF: macrophage migration inhibitory factor; ELISA:enzyme linked immunosorbant assay; VOD: veno-occlusive disease.

Therefore, in continuing efforts to improve the identifica-tion of potential hepatotoxicity issues during lead optimiza-tion, the addition of new, predictive assays to the discoveryarmamentarium is necessary. In vitro systems are an impor-tant component of these new assay system applications. Useof in vitro systems is not new to investigative toxicology orlead optimization processes. Systems such as primary cellcultures, immortalized cell lines, liver slices and whole, per-fused livers are well-established models used for investiga-tive work (Acosta et al., 1985; Butterworth et al., 1989; Ulrichet al., 1995; Guillouzo et al., 1997; Groneberg et al., 2002).Likewise, the use of HepG2 cell lines and primary hepatocytecell cultures to assess potential cytotoxicity are promoted byseveral commercial laboratories. It is the development of up-dated screening paradigms, which combine analysis of datagenerated by new or modified in vitro assays with estab-lished in vivo methods, that may generate additional insightsto help guide the risk assessment/decision-making process.In combination, these assays provide a more well-roundedassessment of potential risk.

This manuscript will present an example of a multi-tierparadigm using in vitro cultures of immortalized cell lines andprimary hepatocyte cultures to assess potential risk for acuteclinical liabilities, and demonstrate how the results from thesesystems are used to help make recommendations regardinglead optimization and the potential for clinical hepatotoxicity(acute liver failure). Further, the use of these in vitro modelsto assess CYP450 enzyme induction potential and biomarkeridentification will also be described.

In Vitro Cell SystemsThe cell-based systems described herein include immortal-

ized cell lines and primary hepatocyte cultures. The immor-talized cell system is comprised of a series of five cell lines uti-lized together as an assay system. These cells were originallyderived from normal human hepatocytes (Pfeifer et al., 1993).The parent cell line has no significant phase I and phase II me-tabolizing capabilities based upon in-house characterization

17

18 DAMBACH ET AL. TOXICOLOGIC PATHOLOGY

studies that examined mRNA expression levels, protein lev-els and protein activities for the major CYP450 enzymes anduridine phosphoglucuronosyltransferase (UDGPT) I and IIisoforms as compared to primary human hepatocyte expres-sion (Andrews et al., 2003). Four “daughter” cell lines werederived from the parent cell line. Each of the daughter celllines has stable expression of an individual human CYP450enzyme under the control of a cytomegalovirus (CMV) pro-moter (Mace et al., 1997). The CYP450 enzyme expressionand activity of these four cell lines are markedly elevated (25-to 90-fold) when compared to primary human hepatocytes(Figure 1). Therefore, high CYP450 activity in combinationwith the low rate of conjugation results in a maximized abil-ity to produce reactive intermediates from test compounds.These CYP450-transfected cell lines have also been shown tometabolize specific substrates and generate metabolite pro-files qualitatively similar to those produced by isolated hu-man microsome and supersome preparations (Andrews et al.,2003). As such, each of these 5 cell lines represent an exag-gerated case of metabolizing enzyme disruption, e.g., over-or underexpression of 1 or more enzymes, which may predicta rare situation in which this would exist clinically, e.g., as aconsequence of genetic polymorphism and/or environmentalfactors.

The primary cell culture system is well-established andconsists of confluent cultures of freshly isolated hepatocytesfrom humans or preclinical species (Acosta et al., 1985;Butterworth et al., 1989; Ulrich et al., 1995; Guillouzo et al.,1997; Groneberg et al., 2002). Primary hepatocyte cultureshave the full complement of metabolizing enzymes, albeitwith inherent donor-to-donor variability, and thus representan assay system that is more representative of hepatocytesin vivo as compared to immortalized cells (Guillouzo et al.,1993).

FIGURE 1.—The steady-state mRNA expression of cytochrome P450isozymes 3A4, 2C9, 2C19, and 2D6 is greatly elevated in CYP450-transfected,immortalized human hepatocyte cell lines (THLE-3A4, THLE-2C9, THLE-2C19, THLE-2D6) as compared to primary human hepatocyte (PHH) cultures.Data is expressed as fold-increases in expression over primary human hepatocyteexpression levels (black bar).

Applications During Lead OptimizationThe following section describes 3 examples of the uses of

these in vitro systems during the discovery lead optimiza-tion process. The data from these assays are used to helpguide decision making with regard to compound rankingearly in the discovery process, to predict the safety “pro-file” of these compounds and generate recommendations forclinical monitoring, and to optimize or develop additionalin vitro or in vivo preclinical screening assays. These assaysystems are also used in a more traditional manner for in-vestigational and mechanistic studies. This latter topic hasbeen extensively covered elsewhere (Acosta et al., 1985;Butterworth et al.,1989; Ulrich et al., 1995; Guillouzo et al.,1997; Groneberg et al., 2002).

Cytotoxicity as a Predictor of Clinical Acute Hepatotoxic-ity: The immortalized cell line series consisting of CYP450enzyme-transfected cells and the non-metabolizing parentalcell line is currently routinely used in-house during the earlydiscovery process (i.e., prior to in vivo testing) to predictthe potential for clinical acute hepatotoxicity, i.e., acute liverfailure. Specifically, this 5-cell-line set is used as a first-tierscreen to assess whether toxicity may occur, whether toxicityis due to the parent compound or an active metabolite, andwhether, in the instance of a toxic parent compound, there isCYP450 detoxification of this toxic compound. Based uponhistorical information, 90% of all drugs are metabolized byCYP450 enzymes (Watkins et al., 1990). Of these, 6 isoforms,3A4, 2C9, 2D6, 2C19, 2E1 and 1A1/2, account for 95% of allCYP450-mediated reactions with 3A4-mediated metabolismaccounting for 65% percent of this total (Zimmerman,1999b). Clinically relevant polymorphisms have been iden-tified for the 2C9, 2C19, and 2D6 isoforms (Fromm et al.,1997; Nebert, 1997; Tanaka, 1999; Poosup et al., 2000; Roses,2000). The 4 CYP450 enzyme isoforms, 3A4, 2C9, 2C19, and2D6, were chosen for use in the immortalized cell cytotoxi-city assay because these enzymes represent 1 that is largelyresponsible for metabolism of drugs (3A4) and 3 with rele-vant polymorphisms that may contribute to toxicity.

The assay was specifically developed to predict the poten-tial for acute direct or metabolism-mediated hepatic necro-sis and not immune-mediated or cholestatic injury. The ra-tionale for this endpoint was due to the fact that nearly50% of all drug-related hepatic damage is hepatic necro-sis (Zimmerman, 1999a), and that modeling of immune-mediated and cholestatic injury is complex and may requirecoculture systems that are not conducive to medium- or high-throughput screening (Guillouzo et al., 1997). In addition,these assessments require minimal compound, i.e., 2 mg,which is an important consideration during the discovery pro-cess when resources to allow substantial compound synthesisare not present.

The basis of the assay is determination of an IC50 valuefor cytotoxicity using a 12-point dose-response curve. Theendpoint evaluated is a decrease in energy status of the cellmeasured by loss of mitochondrial function, i.e., impairedtetrazolium salt reduction (MTS) or decreased ATP levels.This is a sensitive, early indicator of toxicity that is commonto many mechanisms of cellular injury (Marshall et al., 1995;Sun et al., 1997). Confluent cultures are exposed for 20 hoursto compound and dimethyl sulfoxide (DMSO) vehicle. In

Vol. 33, No. 1, 2005 IN VITRO HEPATOTOXICITY SCREENING 19

addition, a known human hepatotoxic drug, perhexiline, andnonhepatotoxic drug, theophylline, are used as positive andnegative controls, respectively.

To evaluate and validate the ability of this assay systemto predict the potential for clinical hepatotoxicity, 679 mar-keted pharmaceuticals were tested. Structure, pharmacology,human toxicity, and pharmacokinetic data were obtained forall of these drugs to achieve chemical and therapeutic targetdiversity and to relate the in vitro results to clinically relevantexposures and outcomes (Andrews et al., 2003). Overall, theresults of the validation study correlated well with clinicaloutcomes. The assay correctly predicted 585/587 nonhepa-totoxic drugs, 15/21 severely hepatotoxic drugs, i.e., “black-box warnings” or withdrawn drugs, and 51/71 variably hep-atotoxic drugs (Andrews et al., 2003). With regard to the 6severe hepatotoxicants not identified in the assay, in all in-stances, the false-negative results were linked to the absencein the assay of the enzyme necessary for metabolism to thereported reactive metabolite. The 20 false-negative predic-tions in the “variable hepatotoxicant” group all representeddrugs that primarily caused cholestasis in humans.

Based on the findings of the validation set, a test articlewith an IC50 value of 50 µM or less in any of the 5 cell linesis interpreted to have an increased potential risk for clinicalhepatic liabilities (Table 1).

In this case, the IC50 values for each of the 5 cell linesare evaluated together to further interpret the contributionof metabolism by CYP450 enzymes to the development oftoxicity. Examples of three potential outcomes related to tox-icity are shown in Figure 2. The first example is perhexilinemaleate, a drug for which toxicity is thought to be medi-ated by the cationic amphiphilic structure of the moleculeand unaffected by CYP450 metabolism (Halliwell, 1997).Danazol is a drug which is metabolized by CYP3A4 leadingto the formation of an active metabolite and is an exampleof bioactivation. The mechanism of danazol hepatic toxic-ity is not clearly understood at this time, but the selectiveinactivation of the CYP3A4 enzyme by its metabolite hasbeen well documented and may contribute toward toxicity(Betz et al., 1981; Konishi et al., 2001). Finally, felbamatemetabolism is an example of a CYP450-mediated detoxifi-cation pathway. Felbamate toxicity is linked to the formationof a toxic metabolite (atropaldehyde) by esterases (Dieck-haus et al. 2002), and the particular sensitivity of humans tothis effect has been attributed, in part, to a decrease in theCYPP450-mediated metabolic pathways when compared topreclinical species (Dieckhaus et al., 2000).

TABLE 1.—Immortalized cell line cytotoxicity assay interpretation criteriaand recommended follow-up action.

Second-tier primaryIC50 value human hepatocyte

(µM) Interpretation Comment cultures

<50 Increased riskfor clinicalhepaticliability

Liability unlesscompound isvery potent orpoorly soluble

Tests compounds ofinterest forimportance ofmetabolizingpathway

>50 Reduced riskfor clinicalhepaticliability

Tests compounds fornon-P450-mediatedtoxicity

FIGURE 2.—Example potential outcomes of the immortalized hepatocyte cellline system with respect to hepatic toxicity. Perhexiline maleate, an inherentlytoxic compound that is not detoxified by cytochrome P450 enzymes (A). Fel-bamate, a toxic compound that is detoxified by all 4 cytochrome P450 enzymesin the assay system (B), and cytochrome P450-3A4-mediated bioactivation ofdanazol (C).

20 DAMBACH ET AL. TOXICOLOGIC PATHOLOGY

The limitations of this assay are considered when evalu-ating the results. These limitations include predictive valuelimited to a single endpoint (hepatocellular necrosis), diffi-culty in separating major and minor metabolic pathways, andlack of sensitivity due to the lack of a specific enzyme neces-sary for metabolism to the reactive metabolite. Recognizingthese limitations, the use of this assay in early discovery is re-stricted to 2 approaches. In the first instance, it is used early inthe discovery process to screen several compounds, e.g., >10,that may be in the same or different chemical class. In thiscapacity the assay serves as a screen to evaluate the potentialfor hepatotoxicity across candidate compounds for lead opti-mization ranking, and across chemical classes for evaluationof potential structure activity relationships (SAR) to toxicity.This screening allows a first assessment of the potential forhepatic liability and for selection of lead compounds; it isnot meant to result in a “go/no-go” decision. In instances inwhich a compound is cytotoxic, the IC50 value is comparedto ED50 values for in vitro efficacy studies as well as in vivotoxicology, and toxicokinetic data, which may have been ob-tained in preclinical species during efficacy testing. Secondly,this assay is part of a larger profiling paradigm, which takesinto account absorption/distribution/metabolism/elimination(ADME) properties and traditional in vivo animal toxicologystudies to develop a well-rounded assessment of the poten-tial for hepatotoxicity for a lead candidate compound. In thisparadigm, primary human hepatocyte cultures from multi-ple donors are used in a manner identical to the immortal-ized cell assay, i.e., IC50 interpretation, as a second tier as-sessment of lead compounds, because primary culture cellshave a more complete set of phase 1 and phase 2 metabo-lizing enzymes (Guillouzo et al., 1993). The use of primaryhepatocytes would therefore give a better assessment of themetabolic route of the drug in a human. In addition to assess-ment in primary human hepatocytes, ADME properties are

FIGURE 3.—In vitro multi-tier hepatotoxicity screening paradigm illustrating the use of an immortalized human hepatocyte cell line assay system followed byassessment in primary human hepatocytes and evaluation of potential metabolites.

also taken into account, such as whether the compound is aCYP450 substrate or inhibitor and whether reactive metabo-lites are formed (Riley and Kenna, 2004). These data are allevaluated in a manner similar to that outlined in the algorithmshown in Figure 3.

The utility of this assessment is to offer guidance both pre-clinically and clinically regarding the potential for hepaticliability, and focus attention on potential hepatic adverse ef-fects that may occur at very low frequencies. The results ofthese in vitro assays are related to both in vitro and in vivo ef-ficacy exposures, and the projected human dose to formulaterecommendations and follow-up monitoring or investigativeplans as necessary.

CYP450 Enzyme Induction: Drug–drug interactions area significant issue not only related to effects on efficaciousexposure levels, but also as related to the potential to causetoxicity (Ajayi et al., 2000; Fuhr, 2000; Rodrigues et al., 2001;Riley and Kenna, 2004). Two classical scenarios are associ-ated with toxicity derived from drug–drug interactions (Liet al., 1997; Ajayi and Perry, 2000). The first involves com-petitive or noncompetitive inhibition of a CYP450 enzymethat is the principal route of metabolism of a drug with inher-ent toxicity. The result is accumulation of a toxic drug, e.g.,concurrent treatment of terfenadine with other CYP3A4 sub-strates leading to prolonged QT intervals and developmentof serious ventricular arrhythmias (Ajayi et al., 2000). Thesecond scenario is typically associated with drug-mediatedinduction of a CYP450, most typically 3A4, leading to en-hanced enzymatic activity and thus increased production ofreactive metabolites, of which there are numerous examples(Li et al., 1997; Fuhr, 2000).

With regard to enzyme induction, the common triggers forexamination of this include a known class liability, character-istic changes in exposure and/or an increase in liver weight

Vol. 33, No. 1, 2005 IN VITRO HEPATOTOXICITY SCREENING 21

with histological changes suggestive of microsomal induc-tion during in vivo animal studies (Haschek and Rousseaux,1998), or as a result of clinical trial toxicokinetic data sug-gestive of enzyme induction. Primary hepatocyte cell cultureshave proven valuable as a screening modality since these cellsexpress phase I enzymes that are capable of being inducedby the appropriate substrate (Guillouzo et al., 1993; Li et al.,1997; Riley and Kenna, 2004). In addition, comparing re-sults from in vitro primary cultures from preclinical speciesto results from preclinical in vivo toxicokinetic studies allowsevaluation of in vitro–in vivo correlations. If a correlation ispresent, it can then be used for screening purposes, so thatcross-species comparisons can then be made using primaryhepatocyte cell cultures, e.g., rat versus human, and thereforeimprove the risk assessment decisions regarding the potentialissues in humans (Ballet, 1997).

The culture methods for CYP450 enzyme induction inprimary hepatocyte cultures has been described previously(Guillouzo et al., 1993; Maurel, 1996; Li et al., 1997;LeCluyse, 2001; Venkatakrishnan et al., 2001). With regardto endpoint measurements, evaluation of a combination ofchanges in CYP450 enzyme mRNA levels and enzyme ac-tivity will give the most useful information for interpreta-tion (Wrighton et al., 1993; Rodriguez et al., 2001; Perezet al., 2003). The major limitations of these assays are avail-ability of acceptable human donor material and the inherentpolymorphic variability in enzymatic activity across donors.Therefore, assays are typically conducted using cells from atleast 2, but preferably 3, separate donors along with a knownCYP450 isoform-inducing drug as a positive control.

Biomarker Identification Using In Vitro Systems: Usefulbiomarkers may include not only traditional biofluid end-points, i.e., clinical pathology parameters, but also in vitropredictive assays, such as those described above, that signalthe potential for a liability and that have been validated using

FIGURE 4.—An algorithm for the use of in vitro cell lines to identify potential biomarkers of hepatotoxicity illustrating concurrent sampling for transcriptionaland protein profiling.

clinical data. In vitro assay systems may also be useful inbiomarker identification using transcriptional and proteomicprofiling, not only for target-specific investigative applica-tion, but on a more global predictive level (Waring et al.,2001; Kramer et al., 2002, 2003). Thus, biomarkers iden-tified during the discovery process can be used to developspecific in vitro screening systems, to optimize or enhancecurrent in vitro screening systems, and in vivo in preclinicalspecies or as bridging biomarkers.

In the example described here, the immortalized cell lineoverexpressing CYP450-3A4 was used to identify more sen-sitive and/or specific markers of cytotoxicity. These identifiedbiomarkers potentially could be applied to enhance the ac-curacy of prediction by further delineating the categories ofhepatotoxicants, i.e., severe versus variable, as well as sug-gest modes of action for toxicity.

The CYP450-3A4 overexpressing cell line was chosen,as mentioned earlier, because this CYP450 enzyme is re-sponsible for greater than 60% of all drug metabolism. Inbrief, the study design was to identify a diverse group ofclasses of structurally and pharmacologically related com-pound pairs that consisted of a known human hepatotoxicdrug and a known nonhepatotoxic drug. The rationale wasthat such a comparison would allow the pharmacologically-mediated gene changes to be subtracted from the changesassociated with the hepatoxin. A decrease in cellular ATPlevel was used as an indicator of early cytotoxicity in theselection of exposure levels. As a consequence, morpholog-ical changes associated with necrosis were never achieved.In the initial experiments, cultures were exposed to drugs for3 time points, 4, 8, and 20 hours, and the dose used was theIC50 value determined in the cytotoxicity model. All non-hepatotoxic compounds were dosed at 50 µM.

The endpoints for biomarker assessment included bothchanges in transcriptional profiles and protein levels in theconditioned media (Figure 4). This dual platform approach

22 DAMBACH ET AL. TOXICOLOGIC PATHOLOGY

allowed for evaluation of transcriptional and translational re-lationships, as well as to give a broader palette of potentiallyuseful biomarker candidates. The results presented here arefrom a test-set comparison of the thiazolidinedione (TZD)PPARγ agonist compounds pioglitazone and troglitazone.Pharmacologically, pioglitazone is approximately 100-foldmore potent than troglitazone; however only troglitazone hasbeen associated with significant hepatotoxicity and death(Lebovitz, 2002; Lee, 2003). These two compounds havegreater than 90% structural similarity. In the in vitro immor-talized hepatocyte cell line assay for cytotoxicity, pioglita-zone never achieved an IC50 (maximal dose 350 µM) and sois interpreted as nontoxic, while the IC50 for troglitazone was∼34 µM in the nonmetabolizing parental cell line and in eachof the CYP450 overexpressing cell lines, i.e., interpreted aspotentially hepatotoxic.

Changes in transcriptional expression were examined us-ing unsupervised and supervised methods. In the unsuper-vised method, the overall change in the entire expressionprofile for each compound was compared to the overall ex-pression changes induced by the vehicle, DMSO, using re-gression analysis. In addition to pioglitazone and troglita-zone, 2 other TZD drugs, ciglitazone and rosiglitazone, werealso compared. From this global gene expression analysis,which does not discriminate pharmacologically induced genechanges from those associated with injury, compounds can beranked by which overall gene profiles look most like DMSO,i.e., R2 value near 1.0 (Figure 5). Based upon this assess-ment, the safety ranking order for the predicted developmentof hepatotoxicity would be DMSO, rosiglitazone, pioglita-zone, ciglitazone, and troglitazone. Thus, an unsupervisedassessment of the entire expression profile of compounds in asimilar structural and/or pharmacological class may be help-ful in ranking compounds, e.g., when attempting to identifyoptimal lead compounds. This approach can be used on anycell line or from organs obtained from preclinical samples.However, this approach is not useful alone and is best con-sidered with all other in vitro and in vivo efficacy/toxicitydata and any other literature related to a drug class, if thatinformation exists.

Supervised assessment of gene expression changes canbe performed in a variety of ways. In this example, at-test was performed, which compared troglitazone-inducedgene changes versus combined gene changes associated withboth pioglitazone and DMSO treatment. Such a compari-son should separate background and pharmacologically in-duced gene changes from those associated with troglitazone-induced toxicity. Using the t-test comparison (p < 0.001),40 genes were identified which were significantly associ-ated with troglitazone treatment after 8 hours. Principal com-ponent analysis (PCA) using these 40 genes clearly segre-gated the 3 treatments (Figure 6). Further, comparison ofadditional TZD drugs, ciglitazone and rosiglitazone, and 3proprietary compounds (A, B, C), showed a clear segre-gation of compounds and demonstrated which compoundswere more “like” DMSO or troglitazone (Figure 6). Com-parison of the supervised gene expression ranking to DMSOand the known clinical outcomes shows good concordance(Gale, 2001) (Table 2).

Thus, with regard to utilizing these results for risk assess-ment decisions, they may be used to help rank compounds, to

TABLE 2.—Comparison of unsupervised transcriptional profile ranking toclinical outcomes.

Drug Clinical outcomea Gene profile rankb

Rosiglitazone No/rare hepatotoxicity 1Pioglitazone No/rare hepatotoxicity 2Ciglitazone Elevated liver function tests 3Troglitazone Hepatotoxicity 4

aGale (2001); Leibowitz (2002); Lee (2003).b1 = profile most similar to DMSO.

identify specific gene changes to further evaluate the mecha-nistic potential of off-target or exaggerated pharmacologicalactivity, and for biomarker identification. As with unsuper-vised analysis data, these findings are used to help guidedecisionmaking and further investigative work and not as a“go/no-go” determinant.

Concurrent identification of potential protein biomarkersof hepatotoxicity from the same experimental system used fortranscriptional profiling allows for efficient use of resources,reduces inherent interexperiment variability, and, most im-portantly, is a more appropriate comparison of compound-induced changes at both the transcriptional and transla-tional levels. The use of multidimensional packed capillaryhigh performance liquid chromatography coupled to tandemmass spectrometry (LC/LC/MS/MS) allows for simultane-ous identification of large numbers of soluble proteins froma relatively small volume of culture medium (Gao et al.,2004). Using this technology, 2 proteins, 14-3-3 zeta andmacrophage migration inhibitory factor (MIF), were highlyassociated with troglitazone treatment versus pioglitazoneand DMSO treatment in both immortalized and primary hep-atocyte cultures. The identity of these proteins was confirmedusing Western blot and enzyme-linked immunosorbant assay(ELISA) analyses (Gao et al., 2004). Changes in 14-3-3 zetaprotein levels did not correlate with changes in transcrip-tional expression levels, thus demonstrating the usefulnessof combined protein and transcriptional profiling for efficientidentification of biomarkers.

The use of in vitro systems for biomarker identification hasseveral major limitations. This is especially true for the useof immortalized cell lines, which are genotypically and phe-notypically different from organ systems (Riley and Kenna,2004). Likewise, these cultures represent single-cell systems,so can only model events that directly affect the cells beingevaluated. Finally, the assay conditions are highly unnaturalwhen compared to in vivo conditions, and the type of as-say, e.g., cytotoxicity, biases all other results. However, invitro systems are used for reasons of practicality and repro-ducibility, and when the limitations of these systems are takeninto consideration with regard to interpretation, and post hocanalyses are used to verify the results, such systems can beutilized. In the example described here, further characteriza-tion of the identified biomarkers has included evaluation inprimary human hepatocytes and in nonhepatocyte cell culturesystems, as well as evaluation in animal models of hepaticinjury and human biofluids. These technologies offer greatpower and promise for the identification of biomarker genesand proteins. However, the characterization and validation ofthese potentially new biomarkers requires extensive time andresources, and this should be a consideration when concep-tualizing the “goal” of biomarker identification.

Vol. 33, No. 1, 2005 IN VITRO HEPATOTOXICITY SCREENING 23

FIGURE 5.—Unsupervised regression analysis of transcriptional profiles from immortalized cell lines exposed to the thiazoledinedione (TZD) drugs, pioglitazone,rosiglitazone, ciglitazone, and troglitazone, compared to dimethyl sulfoxide (DMSO) vehicle (A) shows quantitative differences among the 4 drugs that can be usedto rank compounds for safety, i.e., profiles most “like” DMSO (B).

24 DAMBACH ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 6.—Principal component analysis (PCA) using a troglitazone-induced hepatotoxicity gene profile, made up of 40 genes significantly associated withtroglitazone treatment, demonstrates a distinct separation from pioglitazone- and DMSO-related changes (A). Further comparison using the troglitazone-inducedhepatotoxicity gene profile to additional thiazoledinedione drugs and proprietary compounds demonstrates ability to segregate compounds for safety rankingpurposes (B).

CONCLUSIONSExperimental in vitro modeling systems have utility in

predictive toxicology profiling, identification of potentialbiomarkers and in mechanistic studies as long as the assump-tions and limitations of the systems are realized. The purposeof the modeling system should be rigorously defined and themodel endpoints validated based on real clinical outcomesfor predictive assays or outcomes that have a mechanisticbasis.

To maximize the predictive value, data obtained from thesesystems should not stand alone, but should be viewed in the

context of a multitiered approach and assessed in combinationwith other toxicology, efficacy, and toxicokinetic parametersincluding traditional in vivo studies. Finally, care must betaken to relate these results to in-life or projected human ex-posures for efficacy and toxicity. The combination of resultsfrom well-validated in vitro systems, new technologies andmore traditional toxicity studies will likely result in more ro-bust risk assessment and decision-making. Specifically, thesesystems are used to identify potential issues and thereby at-tempt to minimize these risks during the discovery lead opti-mization process, as well as understand the relevance of this

Vol. 33, No. 1, 2005 IN VITRO HEPATOTOXICITY SCREENING 25

risk to humans. By “flagging” the potential liabilities earlyon in discovery and attempting to understand their relevance,more useful recommendations and guidance with regard tofollow-up studies or monitoring during the development pro-cess may be possible.

ACKNOWLEDGMENTS

Appreciation is expressed to Dr. Michael Neubauer forperforming the unsupervised and supervised methods for theanalysis of the transcriptional profiles and to Drs. Jun-HsiangLin and Shen-Jue Chen for their work in overseeing and com-pleting the expression profiling experiments.

REFERENCES

Acosta, D., Sorensen, E. B., Anuforo, D. C., Mitchell, D. B., Ramos, K., Santone,K. S., and Smith, M. A. (1985). An in vitro approach to the study of targetorgan toxicity of drugs and chemicals. In Vitro Cell Develop Biol 21, 495–504.

Ajayi, F. O., Sun, H., and Perry, J. (2000). Adverse drug reactions: a review ofrelevant factors. J Clin Pharmacol 40, 1093–101.

Andrews, B., Chen, S., Zhu, M., Moulin, F., and Flint, O. (2003). Evaluation ofcytochrome P450-mediated hepatotoxicity using a high throughput roboticsystem. Drug Metabol Rev 35, 413.

Ballet, F. (1997). Hepatotoxicity in drug development: detection, significance,and solutions. J Hepatol 26, 26–36.

Betz, G., Miller, H. H., and Hales, D. B. (1981). Actions of danazol in vivoon cytochrome P-450 and steroidogenic enzymes in rat testis and livermicrosomal preparations. Am J Obstet Gynecol 141, 962–72.

Bissell, D. M., Gores, G. J., Laskin, D. L., and Hoofnagle, J. H. (2001). Drug-induced liver injury: mechanisms and test systems. J Hepatol 33, 1009–13.

Butterworth, B. E., Smith-Oliver, T., Earle, L., Loury, D. J., White, R. D.,Doolittle, D. J., Working, P. K., Cattley, R. C., Jirtle, R., Michaleopoulos,G., and Strom, S. (1989). Use of primary cultures of human hepatocytesin toxicology studies. Cancer Res 49, 1075–84.

Dieckhaus, C. M., Miller, T. A., Sofia, R. D., and Macdonald, T. L. (2000). Amechanistic approach to understanding species differences in felbamatebioactivation: relevance to drug-induced idiosyncratic reactions. DrugMetab Dispos 28, 814–22.

Dieckhaus, C. M., Thompson, C. D., Roller, S. G., and Macdonald, T. L. (2002).Mechanisms of idiosyncratic drug reactions: the case of felbamate. ChemBiol Interact 142, 99–117.

Fromm M. F., Kroemer, H. K., and Eichelbaum, M. (1997). Impact of P450genetic polymorphism on the first-pass extraction of cardiovascular andneuroactive drugs. Adv Drug Deliv Rev. 27:171–99.

Fuhr, U. (2000). Induction of drug metabolizing enzymes. Clin Pharmacokinet38, 493–504.

Gale, E. A. (2001). Lessons from the glitazones: a story of drug development.Lancet 357, 1870–5.

Gao, J., Garulacan, L. A., Storm, S. M., Hefta, S. A., Opiteck, G. J., Lin, J-L.,Moulin, F., and Dambach, D. M. (2004). Identification of in vitro proteinbiomarkers of idiosyncratic Toxicol in Vitro 18, 533–41.

Groneberg, D. A., Grosse-Siestrup, C., and Fischer, A. (2002). In vitro modelsto study hepatotoxicity. Toxicol Pathol 30, 394–9.

Guillouzo, A., Morel, F. A., Fardel, O., and Meunier, B. (1993). Use of hu-man hepatocyte cultures for drug metabolism studies. Toxicology 82, 209–19.

Guillouzo, A., Morel, F. A., Langouet, S., Maheo, K., and Rissel, M. (1997). Useof hepatocyte cultures for the study of hepatotoxic compounds. J Hepatol26(Suppl 2), 73–80.

Halliwell W. H. (1997). Cationic amphiphilic drug-induced phospholipidosis.Toxicol Pathol 25, 53–60.

Haschek, W. M., and Rousseaux, C. G. (1998). Hepatobiliary system. In Fun-damentals of Toxicologic Pathology, pp. 127–152. Academic Press, NewYork.

Konishi, H., Takenaka, A., Minouchi, T., and Yamaji, A. (2001). Impairment ofCYP3A4 capacity in patients receiving danazol therapy: examination onoxidative cortisol metabolism. Horm Metab Res 33, 628–30.

Kramer, J. A., Blomme, E. A., Bunch, R. T., Davila, J. C., Jackson, C. J., Jones, P.F., Kolaja, K. L., and Curtiss, S. W. (2003). Transcription profiling distin-guishes dose-dependent effects in the livers of rats treated with clofibrate.Toxicol Pathol 31, 417–31.

Kramer, J.A., and Kolaja, K. L. (2002). Toxicogenomics: an opportunity tooptimise drug development and safety evaluation. Expert Opin Drug Saf1, 275–86.

LeCluyse, E. L. (2001). Human hepatocyte culture systems for the in vitroevaluation of cytochrome P450 expression and regulation. Eur J PharmSci 13, 343–68.

Lee, W. M. (2003). Drug-induced hepatotoxicity. New Engl J Med 349, 474–85.Leibovitz, H. E. (2002). Differentiating members of the thiazolidinedione class:

a focus on safety. Diabet Metabol Res Rev 18, S23–9.Li, A. P., Maurel, P., Gomez-Lechon, M. J., Cheng, L. C., and Jurima-Romet, M.

(1997). Preclinical evaluation of drug-drug interaction potential: presentstatus of the application of primary human hepatocytes in the evaluationof cytochrome P450 induction. Chemico-Biol Interact 107, 5–16.

Mace, K., Aguilar, F., Wang, J. S., Vautravers, P., Gomez-Lechon, M., Gonzales,F. J., Groopman, J., Harris, C. C., and Pfeifer, A. M. A. (1997). Afla-toxin B1-induced DNA adduct formation and p53 mutations in CYP450-expressing human liver cell lines. Carcinogen 18, 1291–7.

Marshall, N. J., Goodwin, C. J., and Holt, S. J. (1995). A critical assessmentof the use of microculture tetrazolium assays to measure cell growth andfunction. Growth Reg 5, 69–84.

Maurel, P. (1996). The use of adult human hepatocytes in primary culture andother in vitro systems to investigate drug metabolism in man. Adv DrugDeliv Rev 22, 105–32.

Nebert, D. W. (1997). Polymorphisms in drug-metabolizing enzymes: what istheir clinical relevance and why do they exist? Am J Hum Genet 60, 265–71.

Olsen, H., Betton, G., Robinson, D., Thomas, K., Monro, A., Koaja, G., Lilly,P., Sanders, J., Sipes, G., Bracken, W., Dorato, M., Van Deun, K., Smith,P., Berger, B., and Heller, A. (2000). Concordance of the toxicology ofpharmaceuticals in humans and in animals. Reg Tox Pharm 32, 56–67.

Perez, G., Tabares, B., Jover, R., Gomez-Lechon, M. J., and Castell, J. V. (2003).Semi-automatic quantitative RT-PCR to measure CYP induced by drugsin human hepatocytes. Toxicol in Vitro 17, 643–9.

Pfeifer, A. M. A., Cole, K. E., Smoot, D. T., Weston, A., Groopman, J. D.,Shields, P. G., Vignaud, J. M., Juillerat, M., Lipsky, M. M., Trump, B.F., Lechner, J. F., and Harris, C. C. (1993). Simian virus 40 large tumorantigen-immortalized normal human liver epithelial cells express hepato-cyte characteristics and metabolize chemical carcinogens. Proc Natl AcadSci USA 90, 5123–7.

Poolsup, N., Li Wan Po, A., and Knight, T. L. (2000). Pharmacogenetics andpsychopharmacotherapy. J Clin Pharm Ther 25, 197–220.

Riley, R. J., and Kenna, J. G. (2004). Cellular models for ADMET predictionsand evaluation of drug-drug interactions. Curr Opin Drug Disc Develop7, 86–99.

Rodriguez-Antona, C., Pareja, E., Gomez-Lechon, M. J., and Castell, J. V.(2001). Cytochrome P450 mRNA expression in human liver and its re-lationship with enzyme activity. Arch Biochem Biophys 393, 308–15.

Rodrigues, A. D., and Lin, J. H. (2001). Screening of drug candidates for theirdrug-drug interaction potential. Curr Opin Chem Biol 5, 396–401.

Roses, A. D. (2000). Pharmacogenetics and the practice of medicine. Nature 15,857–65.

Sun, J., Wang, L., Waring, M. A., Wang, C., Woodman, K. K., and Sheil, A.G. (1997). Simple and reliable methods to assess hepatocyte viability inbioartificial liver support system matrices. Artif Organs 21, 408–13.

Tanaka, E. (1999). Gender-related differences in pharmacokinetics and theirclinical significance. J Clin Pharm Ther 24, 339–46.

Ulrich, R. G., Bacon, J. A., Cramer, C. T., Peng, G. W., Petrella, D. K., Stryd, R. P.,and Sun, E. L. (1995). Cultured hepatocytes as investigational models forhepatic toxicity: practical applications in drug discovery and development.Toxicol Lett 82/83, 107–15.

26 DAMBACH ET AL. TOXICOLOGIC PATHOLOGY

Venkatakrishnan, K., von Moltke, L. L., and Greenblatt, D. J. (2001). Humandrug metabolism and the cytochromes P450: application and relevance ofin vitro models. J Clin Pharmacol 41, 1149–79.

Waring, J. F., Ciurlionis, R., Jolly, R. A., Heindel, M., and Ulrich, R. G. (2001).Microarray analysis of hepatotoxins in vitro reveals a correlation betweengene expression profiles and mechanisms of toxicity. Toxicol Lett 120,359–68.

Watkins P. B. (1990). Role of cytochromes P450 in drug metabolism and hepa-totoxicity. Semin Liver Dis 10, 235–50.

Wrighton, S. A., VanderBraden, J. C., Stevens, L., Shipley, L. A., Ring, B. J.,Rettie, A. E., and Cashman, J. R. (1993). In vitro methods for assessinghuman hepatic drug metabolism: their use in drug development. DrugMetabol Rev 25, 453–84.

Zimmerman, H. J. (1999a). The spectrum of hepatotoxicity. In Hepatotoxicity,2nd ed., pp. 3–10. Lippincott Williams & Wilkins, Philadelphia.

Zimmerman, H. J. (1999b). Hepatic metabolism of foreign compounds. InHepatotoxicity, 2nd ed., pp. 11–40. Lippincott Williams & Wilkins,Philadelphia.