4,775
Views
14
CrossRef citations to date
0
Altmetric
Review Articles

Through a glass, darkly? HepaRG and HepG2 cells as models of human phase I drug metabolism

&
Pages 46-62 | Received 19 Jan 2022, Accepted 03 Feb 2022, Published online: 23 Feb 2022

Abstract

The pharmacokinetic and safety assessment of drug candidates is becoming increasingly dependent upon in vitro models of hepatic metabolism and toxicity. Predominant among these is the HepG2 cell line, although HepaRG is becoming increasingly popular because of its perceived closer resemblance to human hepatocytes. We review the functionality of these cell lines in terms of Phase I protein expression, basal cytochrome P450-dependent activity, and utility in P450 induction studies. Our analysis indicates that HepG2 cells are severely compromised: proteomic studies show that they express few key proteins in common with hepatocytes and they lack drug-metabolizing capacity. Differentiated HepaRGs are more hepatocyte-like than HepG2s, but they also have limitations, and it is difficult to assess their utility because of the enormous variability in data reported, possibly arising from the complex differentiation protocols required to obtain hepatocyte-like cells. This is exacerbated by the use of DMSO in the induction protocol, together with proprietary supplements whose composition is a commercial secret. We conclude that, while currently available data on the utility of HepaRG generates a confusing picture, this line does have potential utility in drug metabolism studies. However, to allow studies to be compared directly a standardized, reproducible differentiation protocol is essential and the cell line’s functionality in terms of known mechanisms of P450 regulation must be demonstrated. We, therefore, support the development of regulatory guidelines for the use of HepaRGs in induction studies as a first step in generating a database of consistent, reliable data.

Introduction

The human liver tumor-derived cell lines HepG2 and HepaRG are widely used in academia and industry: HepG2 is well-established in the prediction of hepatotoxicity, including the potential for drug-induced liver injury (Weaver et al. Citation2017, Citation2020), while HepaRG has, in recent years, become increasingly popular for studies on xenobiotic metabolism, including the prediction of intrinsic clearance (Lübberstedt et al. Citation2011; Bonn et al. Citation2016; Kratochwil et al. Citation2017). Such widespread dependence upon these cell lines raises the question of how closely they reflect primary human hepatocytes and consequently ‘Are these cell lines sufficiently informative to be used for regulatory purposes?’

Immortalized cell lines like HepG2 and HepaRG are intrinsically unlike healthy human hepatocytes, yet their use in drug discovery and development involves assuming that their metabolic activity and gene regulatory networks accurately reflect those of human hepatocytes in vivo. We set out to review the ability of these cell lines to represent human hepatic Phase I metabolism with sufficient reliability and reproducibility for research purposes and acceptance by regulators.

In this context, studies that only measure changes in mRNA expression are of limited value because such changes often do not segregate with protein level and functional activities (Buccitelli and Selbach Citation2020). In the case of P450 induction, it is known that fold changes in mRNA expression frequently fail to accurately reflect changes in enzyme activity (Chu et al. Citation2009). We, therefore, excluded studies in which expression and/or induction were addressed only at the mRNA level.

In considering HepaRG and HepG2 cells as models of human hepatocyte function, this commentary focused on measurable phenotypic characteristics, particularly enzyme activity, evaluating the cell lines in terms of basal activities with diagnostic substrates compared against those of freshly prepared human hepatocytes and inducibility compared against that of cultured human hepatocytes. These characteristics are evaluated in the context of the cell lines’ history, genetic characteristics, and protein expression profiles.

Background

The HepG2 cell line, established by Knowles et al. (Citation1980), was soon shown to be capable of activating cyclophosphamide to genotoxic products, resulting in sister chromatid exchanges, and to exhibit some benzphetamine N-demethylation activity (Dearfield et al. Citation1983). It could hydroxylate 7-ethoxycoumarin, this activity being induced 20- to 30-fold after 3–4 days’ exposure to 3-methylcholanthrene (3-MC, 5 µM) (Dawson et al. Citation1985). No induction was detected using phenobarbital (PB). Subclones with higher P450-dependent activities were subsequently isolated. One of these, HepG2/C3A, exhibited 7-ethoxyresorufin and phenacetin O-deethylase activities consistent with the expression of cytochrome P450 (CYP) 1A2 and responded to 3-MC with up to 40-fold induction (Kelly and Sussman Citation2000). This cell line was patented (Patent Numbers US5290684A, 1994 and US6653105B2, 2003) and used to develop a commercial fluorescence-based assay for CYP1A2 induction; these patents have expired and the HepG2/C3A cell line is available from the American Type Culture Collection (ATCC HB-8065).

The patent-protected HepaRG cell line (PCT/FR02/02391, 2002) was established from a Grade I hepatocarcinoma growing in an adult female with chronic hepatitis C infection. The resulting cultures comprised bipolar oval cells with the capacity to differentiate when treated with dimethyl sulfoxide (DMSO; ∼2% in cell culture medium) and hydrocortisone (50 µM) (Parent et al. Citation2004). The resulting hepatocyte-like cells metabolized phenacetin, tolbutamide, dextromethorphan, and nifedipine with specific activities ∼2–10 pmol/min/mg protein (Gripon et al. Citation2002), while fluorescence-based assays demonstrated 7-ethoxyresorufin O-deethylation (EROD), tolbutamide 4-hydroxylation, testosterone 6β-hydroxylation and chlorzoxazone 6-hydroxyation in differentiated HepaRGs; EROD activity was upregulated by 3-MC (5 µM, 24 h) but testosterone 6β-hydroxylation was not further increased in response to the CYP3A4 inducer rifampicin (25 µM, 72 h) (Aninat et al. Citation2006).

HepaRGs only exhibit these characteristics in the presence of hydrocortisone and high concentrations of DMSO. When thus treated, most of the cells die; those that survive form small clusters of hepatocyte-like cells which organize themselves into well-delineated trabeculae surrounded by biliary epithelial-like cells. The expression of CYP3A4 in the surviving cells is attributable to the presence of the differentiating agents, which are known to upregulate CYP3A4 (El-Sankary et al. Citation2000; Xie et al. Citation2020). This P450 profile is likely to reflect that of inducer-exposed hepatocytes rather than naïve cells, a consideration to keep in mind when evaluating induction studies using HepaRGs. It should also be noted that DMSO inhibits several P450 isoforms including CYP3A4 (Easterbrook et al. Citation2001).

Genetic profile

Different HepG2 stocks have disturbingly variable chromosome complements: one study specifies the karyotype as 55 compared with the normal human karyotype of 46 XX or 46 XY (Wiśniewski et al. Citation2016), while others report karyotypes 49–52 XY or 52–78 XY with numerous duplications, deletions, and rearrangements (Wong et al. Citation2000; Zhou et al. Citation2019). This may reflect the cell line’s evolution over the >40 years since it was established. Many HepG2 stocks do not closely resemble human hepatocytes (Tascher et al. Citation2019), illustrating the importance of obtaining stocks of any cell line from reputable sources (e.g. the ATCC).

A detailed haplotype-resolved and integrated genome analysis of HepG2s acquired from the Stanford ENCODE Product Center for Mapping of Regulatory Regions provides a detailed karyotype (Zhou et al. Citation2019). This emphasizes the line’s many structural and numerical abnormalities and offers a comprehensive analysis of genomic structural features including single nucleotide variants, copy number and ploidy changes, phased haplotype blocks, CRISPR targets, novel retrotransposon insertions, and structural variants including deletions, duplications, inversions, translocations, and complex rearrangements.

Only limited information on the karyotype of HepaRGs is available. They carry a supernumerary (remodeled) copy of chromosome 7 and only have one intact copy of chromosome 22; a t(12:22) translocation leads to monosomy of 12p (Gripon et al. Citation2002), but at least part of chromosome 22 must remain diploid because the cells have been genotyped as CYP2D6*2*9, indicating that they carry two copies of CYP2D6 (Jackson et al. Citation2016). The CYP2D6*2 group of alleles, which is present in almost every population studied, encodes an enzyme with activity resembling that of the reference haplotype, CYP2D6*1. Its several sub-alleles differ from the reference sequence at 2581 C>T and 4181 G>C, leading to an Arg to Cys substitution at amino acid 296 and a Ser to Thr substitution at amino acid 486 (Nofziger et al. Citation2020); it is unclear which sub-allele the HepaRG cell line contains. CYP2D6*9 is characterized by a 3 bp deletion commonly described as g.2616 del AAG (rs5030656) leading to the deletion of Lys 281 (Nofziger et al. Citation2020). Cyp2D6*9 is classified as a reduced function haplotype (https://www.pharmgkb.org/haplotype/PA165948317) because the variant protein it encodes has reduced activity and is expressed at lower levels than the reference haplotype; HepaRGs are therefore predicted to be of CYP2D6 intermediate metabolizer phenotype.

HepaRGs are homozygous for a poor metabolizer allele of CYP2C9 (CYP2C9*2*2). The CYP2C9*2 variant (3608 C>T, rs1799853) has an Arg to Lys substitution at amino acid 144 leading to significantly reduced catalytic activity and decreased clearance of CYP2C9 substrates in vivo (Zanger and Schwab Citation2013). Clinical consequences include an increased risk of bleeding during treatment with anticoagulants such as warfarin, and adverse events in patients treated with sulfonylurea anticoagulants and non-steroidal anti-inflammatory drugs. The allele frequency of CYP2C9*2 is 8–13% in European, Central/South American, Near Eastern, and Latino populations but <3% in other populations (Sangkuhl et al. Citation2021). Only 1–2% of the Caucasian population is homozygous for this variant (Zanger and Schwab Citation2013).

HepaRGs are also homozygous for CYP3A5*3 (Jackson et al. Citation2016). This variant (6986 A>C; rs776746) creates a splicing defect in CYP3A5 intron 3. Leading to reduced expression and enzymic activity, it is associated with reduced metabolism and dose requirements for drugs that are preferentially metabolized by CYP3A5, including tacrolimus and saquinavir (Zanger and Schwab Citation2013).

The homozygous poor metabolizer origin of HepaRGs for both CYP2C9 and CYP3A5 together with their CYP2D6 heterozygosity (one extensive and one poor metabolizer allele) means that they do not represent the majority of the human population, at least in terms of metabolism mediated by these isoforms.

We are not aware of a publicly available genome sequence for HepaRG, but this information would establish how closely HepaRGs resemble human hepatocytes and is key to the informed interpretation of data obtained with this cell line. A genome sequence would also reveal whether novel mutations acquired during tumor development or subsequent in vitro culture are present in P450 promoter regions or transcription factors, e.g. the constitutive androstane receptor (CAR), pregnane X receptor (PXR). Sequencing of multiple cultures would also indicate the rate of genetic drift, as seen in the HepG2 line.

Expression of phase I proteins

Several groups have compared HepG2 and HepaRG proteomes with those of the human liver, either in the form of cultured hepatocytes or tissue biopsies.

Comparing HepG2s, grown either as monolayers or spheroids, with cryopreserved PHHs analyzed as cell suspensions revealed similar findings (Wiśniewski et al. Citation2016; Hurrell et al. Citation2018). Most PHH batches clustered separately from HepG2s, which exhibited similar proteomes in 2D and 3D cultures. All P450 isoforms studied (CYPs 1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 3A4, and 3A5) were readily identified in PHHs but barely detectable in HepG2s, which expressed a strong cancer cell signature, the proteins found at high abundance being those associated with cell proliferation, including components of the DNA replication, transcription, and translation machinery (Hurrell et al. Citation2018).

Focusing on Phase I by comparing HepG2 and other cell line microsomes with commercially acquired human liver microsomes supports these conclusions. The levels of 44/101 xenobiotic-metabolizing enzymes, including 19/24 P450s, were below the limit of detection (BLD) in HepG2 microsomes, the overall expression pattern of HepG2 microsomes being distinct from those of the Hep3B and Huh7 cell lines and PHHs (Shi et al. Citation2018).

In a comparison between differentiated HepaRGs, HepG2s, Upcyte® hepatocytes and cryopreserved primary human hepatocytes (PHHs), Sison-Young et al. (Citation2015) identified a total of 4696 proteins, only 2722 of which were common to all the models evaluated. The overall proteomic profile of HepaRGs was closest to that of cryopreserved PHHs, although these were clearly segregated from the other models examined. This relationship was maintained when the analysis was restricted to 50 Phase I, 51 Phase II, and 27 Phase III proteins. Looking specifically at CYPs 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4, 3A5, 4A11, 4F11, 4F12, 4V2, and 20A1, the expression of CYP3A4 in HepaRGs was 2.5x that in cryopreserved PHHs, the only case where a P450 was overexpressed in a cell line. Western immunoblotting confirmed that CYP1A2 expression was BLD in both HepaRGs and HepG2s; CYP2D6 was barely detectable in HepaRGs but not at all in HepG2s. Strong expression of CYP2E1 and CYP3A4 was detected in HepaRGs differentiated in house, but not in HepG2s.

HepaRGs are now available in a cryopreserved, pre-differentiated format (HPR116), which may help to reduce study-to-study variability. These cryoHepaRGs lack metabolic competence when first thawed but recover if maintained in culture for 7–10 days after thawing; they have been used for the assessment of basal P450-dependent activities (Jackson et al. Citation2016). Comparing HPR116 with PHHs and HepG2s, Tascher et al. (Citation2019) found marked differences in the abundance of many proteins between HepaRGs and PHHs (1790 proteins) as well as between HepaRGs and HepG2s (1598) and HepG2s and PHHs (1646). While the expression of structural proteins was generally similar HepaRGs and PHHs, there were exceptions, particularly associated with contractility, trafficking, cell-cell communication, polarity, and morphogenesis; however, the majority of Phase I and II proteins exhibited comparable expression. The differences observed were attributed to the HepaRG trans-differentiation process and an altered balance between senescence and proliferation typical of transformed cells.

Hammer et al. (Citation2021) report a detailed evaluation of protein expression in HepaRGs. Targeted proteomic analysis of HepaRGs acquired as undifferentiated stocks and differentiated in house, Wistar rat liver, C57BL/6 mouse liver, PXR/CAR-humanized mouse liver, FRG®-KO mouse liver populated with human hepatocytes and PHHs was compared with historical human liver biopsy results (summarized in ), revealing very low levels of CYPs 1A2, 2B6, 2C8, 2C9, 2D6, and 2E1 in differentiated HepaRGs compared with PHHs. The level of CYP3A4 in HepaRGs resembled that in PHHs, possibly due to the inducing effect of DMSO. CYP2B6 was expressed at levels lower than those in PXR/CAR humanized mouse liver, humanized FRG®-KO mouse liver, and PHHs but resembling those of the equivalent proteins, CYP2B1 and Cyp2b10, in uninduced wild-type Wistar rat and C57BL/6 mouse liver.

Table 1. Proteomic analysis of cytochrome P450 expression in HepaRG cultures, cryopreserved PHHs and human liver biopsies.

One challenge in proteomic analysis of P450s in hepatocytes and cell lines is that little is known about the expression of the key nuclear receptors (NRs) PXR and CAR at the protein level. They are not addressed in the proteomic studies discussed above, and a lack of specific antibodies means that their expression has not been characterized immunochemically, either immunohistologically or by immunoblotting.

Basal P450-dependent activities

Consistent basal enzyme activities comparable to those of the human liver are a key feature of any model of human drug metabolism. In the context of Phase I metabolism, the model’s basal P450-dependent activities should resemble those of freshly prepared PHHs.

P450-dependent activities tend to be close to or below the limit of detection in HepG2s, which are sometimes used as a negative control against which to compare other cell types because of their very limited metabolic activity (Kratochwil et al. Citation2017). Only very occasionally is measurable activity reported, and this is usually not replicated in other studies using the same substrates. Only for CYP3A4 are there several reports of measurable activity, and even then the activities are very low, midazolam 1′-hydroxylation in HepG2s being, at best, ∼100-fold lower than in monolayer cultured PHHs. Overall, therefore, HepG2s do not express measurable basal levels of P450-dependent xenobiotic metabolism.

compares basal P450-dependent activities in HepaRGs with those of PHHs. This comparison should, ideally, be made using freshly prepared PHHs, but in practice most investigators use cryopreserved stocks assayed immediately after thawing (possibly with a brief recovery period in suspension before assay) or cultured for a period before activities are measured. , therefore, includes studies undertaken using both fresh and cryopreserved cells. It is restricted to studies which (i) address two or more of the key models (HepG2s or HepaRGs grown in monolayer culture, cultured PHHs or PHH suspensions); (ii) measure activities using diagnostic substrates applied directly to intact cells; (iii) use liquid chromatography/mass spectrometry (LC/MS) to quantify either substrate depletion or metabolite production; and (iv) report specific activities (as pmol/min/mg protein or pmol/min/106 cells). Studies that do not report specific activities but only make internal comparisons between two systems may provide additional supportive evidence but are not tabulated because their results cannot be compared with those of other investigators. Furthermore, assay data are often presented in graphical rather than tabular form making it impossible to compare studies statistically; however, some qualitative conclusions can be drawn ().

Table 2. Basal P450-dependent activities in HepG2 and HepaRG cultures compared with PHHs.

Table 3. Summary of basal P450-dependent activities in HepG2 and HepaRG cultures.

Even when all four criteria are met there is enormous variability both within and between the reported data. Regarding individual human preparations, this could be partly due to the biological variation but variation within a particular cell line is likely to be due to the use of different culture systems, differentiation protocols, and assay methodologies; indeed, different activity values have been reported in a single study depending upon which of two different substrate cocktails was used (Anthérieu et al. Citation2010).

P450-dependent activities are generally higher in differentiated HepaRGs than in HepG2s, even the HepG2/C3A subclone (Nelson et al. Citation2017), and can resemble those in PHHs cultured in monolayer format for at least 48 h. When compared with primary hepatocyte suspensions, however, both HepaRGs and monolayer cultured PHHs exhibit much lower, sometimes barely detectable, activities. The only exception is that HepaRGs consistently exhibit CYP3A4-dependent activities at least as high as in monolayer cultured PHHs and sometimes even higher, whether measured as midazolam 1′-hydroxylation or testosterone 6β-hydroxylation; indeed, this is the only case in which a P450-dependent activity observed in a cell line resembles that of PHH suspensions. Even in this case, however, HepaRG activities tend to be toward the lower end of the range when large numbers of PHH batches are included (Jackson et al. Citation2016; Ramaiahgari et al. Citation2017). In general, therefore, apart from CYP3A4, HepaRGs exhibit at best moderate P450-dependent activities compared with PHHs, even allowing for the considerable variability between PHH cultures.

Most studies of P450-dependent metabolism in HepaRGs only measure activities at a single time after the initiation of differentiation, usually in comparison with monolayer cultured PHHs also assayed at a single time after plating. Such studies are limited because cell lines and primary cultures are dynamic systems: for example, P450-dependent activities drop by 10- to 100-fold when PHHs are assayed in situ after 4- or 48-hr adhesion time (Ulvestad et al. Citation2013).

Only one study has explicitly considered the time course of P450-dependent metabolic capability in HepaRGs, addressing three timepoints during the differentiation process (‘Days 0, 7, and 14,’ equivalent to days 14, 21, and 28 after shipping) compared with cultured PHHs over the same time course starting on day 3 of culture (Lübberstedt et al. Citation2011). In both culture systems CYP1A2 activity (measured as EROD) was stable over this culture period, being slightly lower in HepaRGs than cultured PHHs but much less variable. The activity was much higher than that of cryopreserved PHHs assayed immediately after thawing, raising the possibility that the activity detected may reflect artefactual upregulation of CYP1A1 and/or 1A2 due to dedifferentiation in culture. HepaRGs and monolayer cultured PHHs exhibited similar CYP2C9, CYP2C19, and CYP2D6 activities (diclofenac 4-hydroxylation, S-mephenytoin hydroxylation, and bufuralol 1-hydroxylation) on day 0; these activities were, however, much lower than those in cryopreserved PHHs assayed in suspension immediately after thawing. Over 14 days of differentiation CYP2C9, 2C19, and 2D6 activities in HepaRGs were maintained or increased slightly, whereas activities in monolayer cultured PHHs declined rapidly so that by the end of the differentiation period the activities in HepaRGs were generally higher than those in cultured PHHs. In contrast with all the other P450-dependent activities, CYP3A4 activity (testosterone 6β-hydroxylation) was considerably higher in HepaRGs than in either cultured PHHs or cryopreserved PHHs assayed in suspension, activity in the latter being higher than in cultured PHHs but still lower than in HepaRGs.

It is important to remember that the presence of DMSO is essential to the maintenance of P450-dependent activities in HepaRGs. Removal of DMSO from the culture medium alters P450 expression and activity within 24 h (Kanebratt and Andersson Citation2008a), although it is unclear whether this is due to loss of the P450-inducing effect of DMSO or general dedifferentiation of the cells in the absence of its differentiating stimulus.

Regulation of P450 expression and activity

The regulation of P450-dependent metabolism, a primary function of hepatocytes, influences both xenobiotic metabolism and susceptibility to hepatotoxins. The most important pathways of P450 regulation are mediated by the PAS domain aryl hydrocarbon receptor (AhR) and the NRs PXR and CAR.

A wide range of high and low-affinity ligands can activate AhR, initiating a pathway that has been conserved through evolution and is functional in most mammalian cell types as well as in tumor cell lines. The activity of this pathway, exemplified by the induction of CYP1A1/1A2 by polycyclic aromatic hydrocarbons, therefore does not reflect a differentiated hepatocyte phenotype.

Regulation via PXR occurs in several tissues including the liver and gastrointestinal tract (Koutsounas et al. Citation2013). The mechanism of action of PXR resembles that of conventional NRs, such as the steroid hormone receptors, except that, instead of binding a narrow range of ligands with very high affinity, it binds a wide range of ligands with relatively low affinity (Stanley et al. Citation2006). This pattern of ligand responsiveness may have evolved to allow PXR to deal with the many xenobiotics to which organisms are exposed.

The liver-specific receptor CAR can be activated both by agonists [e.g. 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO)] and inverse agonists (e.g. androstanol) which inhibit its function. Other inducing agents, including PB, regulate CAR by indirect mechanisms involving protein phosphorylation and dephosphorylation (McMahon et al. Citation2019; Negishi et al. Citation2020). It is important that studies involving functional analysis of CAR investigate both pathways of regulation.

Studies of P450 induction in cultured liver cells have been driven by the need for reliable models to predict drug-drug interactions, especially those which occur because of PXR/CAR-mediated induction of CYP3A4 and CYP2B6. Most studies addressing the induction of CYP2B6 and CYP3A4 at the enzymatic level have been conducted in HepaRGs, sometimes in comparison with PHHs.

This literature is difficult to interpret because of study-to-study inconsistencies in the induction protocols used: when undertaking this review we were unable to identify any two studies in which the same inducing agent, concentration, and duration of exposure were used. In addition, many investigators do not report the basal expression levels against which fold inductions are calculated; the fold induction which is considered meaningful is often not defined; and the activities measured are often close to the limit of detection of the assays used, meaning that even induced activities may not be pharmacologically significant. Furthermore, many studies are conducted at only one concentration of the inducing agent, meaning that a dose-response curve cannot be constructed and making it even more difficult to compare studies. Finally, it is important to note that PXR/CAR ligand-binding affinities do not necessarily predict maximal induction ratios (Stanley et al. Citation2006; Scheer et al. Citation2008).

P450 induction in HepG2 cultures

P450 protein expression in HepG2s exhibits only limited responsiveness to P450 inducing agents. Consistent with their origin from a hepatoblastoma, they respond to AhR agonists with upregulation of immunochemically detectable CYP1A2, phenacetin O-deacetylation, and EROD; Choi et al. (Citation2015) also report slight induction (about 2-fold) of bupropion hydroxylase by PB (1 mM for 48 h) and testosterone 6β-hydroxylase activity by both PB (1 mM for 48 h) and rifampicin (50 µM for 48 h), respectively, but it is unclear against what basal activities this induction was measured, making it difficult to compare their results with those of other investigators. Others have found expression levels of CYP3A4 which were barely detectable by immunoblotting and no evidence of induction by CITCO or rifampicin in HepG2s (Harmsen et al. Citation2008).

Only two studies addressing PXR- and CAR-mediated induction in HepG2s met our inclusion criteria. Phenobarbital had no effect on midazolam 1′-hydroxylation, which was 1.1× control following treatment at 500 µM for 72 h (Gerets et al. Citation2012). Similarly, rifampicin had very little inductive effect in HepG2s: Berger et al. (Citation2016) report marginal induction of omeprazole metabolism (1.3-fold) and midazolam 1′-hydroxylation (1.3 and 1.7-fold, respectively, after 72 h at 20 µM (Berger et al. Citation2015), while Gerets et al. (Citation2012) report no change in midazolam 1′-hydroxylation in microsomal preparations after 72 h at 25 µM. In each of these studies, rifampicin and PB had readily detectable effects on PHHs and HepaRGs (). At the present time the mechanism of the reduced CAR activity in HepG2 cells is unknown; it may be due to genetic or epigenetic changes which attenuate the signaling cascade(s) involved.

Table 4. Studies comparing induction of CYP2B6 and CYP3A4-dependent activities in PHHs and HepaRG cultures.

CYP3A4 induction in HepaRG cultures

summarizes studies addressing the induction of midazolam 1′-hydroxylation (CYP3A4) by rifampicin in HepaRGs, which indicate fold induction values resembling those in PHHs. It is not possible to evaluate the relationship between fold induction and rifampicin concentration from these experiments, but a dose-response study using four concentrations of rifampicin provides some evidence for concentration dependence (Kaneko et al. Citation2009). There was, however, wide batch-to-batch variability. In some batches the induction response tailed off at higher concentrations; thus, the EC50 (4.3×10−3 mM) was outside the concentration range over which induction was determined to be dose-dependent. Kanebratt and Andersson (Citation2008b) found HepaRGs to be much more sensitive to rifampicin, deriving an EC50 of 1.2×10−4 mM (36-fold lower) for induction of midazolam 1′-hydroxylation; this may reflect the fact that their study did not include concentrations at which the inductive response tailed off. Fold induction of testosterone 6β-hydroxylation in HepaRGs exhibited signs of dose-responsiveness with rifampicin induction but plateaued above 100 nM with hyperforin (Jackson et al. Citation2016).

Several studies have also considered the induction of CYP3A4 via indirect activation of CAR. The fold values obtained in single-dose studies using PB are again highly variable (). This lack of consistency makes it difficult to evaluate how PB induction of CYP3A4 in HepaRGs compares to that in PHHs, especially as many studies do not report the basal activities against which fold induction is being calculated.

Kaneko et al. (Citation2009) characterized the effects of different concentrations of PB and carbamazepine on the induction of CYP3A4. There was enormous batch-to-batch variation in the response: one of the four batches tested responded strongly to both PB and carbamazepine whereas another exhibited practically no induction with either of these compounds even at the highest doses tested. The EC50s derived from these data were 28.7 mM for PB and 3.8 mM for carbamazepine, based on experiments in which hypothetical maximal induction was never reached; the uncertainty underlying these values is reflected in the coefficients of variation (88.6% for PB and 198.3% for carbamazepine).

Taken together, these studies suggest dose-dependent induction of CYP3A4 by indirect activators of CAR, but the widely variable results highlight concerns about batch-to-batch variability in HepaRGs.

CYP2B6 induction in HepaRG cultures

In studies addressing the effects of CAR activators on CYP2B6-mediated activities in HepaRGs, neither CITCO nor phenytoin had more than a slight effect on bupropion hydroxylation, while the effects of PB were extremely variable. These data suggest that the function of CAR is, at best, compromised in HepaRGs (). The changes observed do not reflect observations in patients treated with CAR activators, such as PB and phenytoin, which are known to cause significant changes in drug exposure (Khoo et al. Citation1980).

Studies using rifampicin suggest that CYP2B6 can be regulated to a limited extent by PXR in HepaRGs; where a comparison was made the values for HepaRGs were in the same range as those detected in PHHs (), although the very low constitutive expression of CYP2B6 in HepaRGs should be borne in mind. When background expression is very low, even large fold changes are unlikely to lead to measurable changes in functional activity.

One approach to circumvent the activity of CAR in HepaRGs is to engineer the cells to constitutively overexpress CAR. Van der Mark et al. (Citation2017) achieved this using a lentiviral vector encoding CAR driven by the murine phosphoglycerate kinase 1 promoter. HepaRG-CAR cells exhibited increased activity of both CAR target and non-target P450s and increased clearance of warfarin and prednisolone, although mRNA induction ratios following treatment with omeprazole or CITCO were unchanged. Interestingly, CYP3A4 mRNA induction by rifampicin was ablated in HepaRG-CAR cells, consistent with the idea that CYP3A4 is fully induced in HepaRGs without induction. HepaRG-CAR cells are presented as a more physiologically relevant model for studies on differentiation and metabolism in this cell line without the need for exposure to DMSO.

Effects of rifampicin on other P450-dependent activities in HepaRGs

Various studies have looked more widely at the effects of rifampicin on P450-dependent activities in HepaRGs, considering activities beyond those mediated by CYP2B6 and CYP3A4. The activities reported generally resembled those in PHHs, although in a few cases greater fold induction values were observed in HepaRGs (). This reflects one of the limitations of cultured cells which express low basal levels of particular P450 isoforms: even where high induction ratios are reported the resulting enzyme level or activity may still be very low. This may also help to explain why the pattern of induction seen for a particular P450 often depends upon the substrate used to measure its activity: thus, marginal, if any, induction of CYP2C19 is observed in either PHHs or HepaRGs when measured as mephenytoin 4-hydroxylation (Lübberstedt et al. Citation2011), but around 4-fold induction is observed using omeprazole as substrate (Berger et al. Citation2016).

Table 5. Rifampicin-induced changes in P450-dependent activities in PHHs and HepaRG cultures.

Use of NR-knockout HepaRGs to elucidate induction mechanisms

Knockout (KO) approaches have been used to characterize the contribution of PXR to the induction of CYP2B6 and CYP3A4 mRNAs in HepaRGs. Note that this section is an exception to our policy of including only phenotypically anchored results: protein expression and enzyme activity are rarely if ever, evaluated in studies of this kind.

A zinc-finger targeted PXR-KO HepaRG cell line has been shown to be refractory to rifampicin induction and has greatly decreased sensitivity to CYP3A4 induction by dual activators of PXR and CAR (PB, phenytoin, and carbamazepine), possibly because CYP3A4 is already maximally upregulated in this model. The slight residual activity seen with these agents was thought to be due to upregulation of CYP3A4 via CAR; consistent with this, the small CYP3A4 inductive response to CITCO was maintained in the PXR-KO, although this is difficult to reconcile with evidence suggesting that HepaRGs lack CAR functionality. Artemisinin did not upregulate CYP3A4 in PXR-KO cells, suggesting that artemisinin is PXR-specific for CYP3A4 induction in this context, although it did exhibit some activity in CYP2B6 induction (Williamson et al. Citation2016).

A study using PXR-KO, CAR-KO, and double PXR/CAR-KO HepaRGs (Preiss et al. Citation2021) is summarized in . The PXR-KO yielded results consistent with those in PXR-KO mice, in which CYP3A expression (Cyp3a11 in mice) undergoes compensatory upregulation, with no scope for further induction in response to rifampicin (Scheer et al. Citation2008), and as expected no induction occurred in the CAR-KO cultures treated with CITCO. However, basal CYP3A4 mRNA expression was decreased rather than increased in the double PXR/CAR-KO, and a mixture of effects was seen in CAR-KOs and PXR/CAR-KOs, possibly because CAR function is already compromised in HepaRGs, making the effects of a knockout difficult to interpret. In this study mRNA levels were expressed relative to those in control HepaRGs, in which levels of expression may have been very low, so one cannot conclude absolute values from the results. Models of this kind facilitate the study of PXR/CAR-independent mechanisms of induction, although it is impossible to be sure that such pathways function in HepaRGs in the same way as in hepatocytes.

Table 6. Cytochrome P450 mRNA expression and induction in Nuclear Receptor Knockout HepaRGs.

HepaRGs have also been used in knockdown studies to elucidate the role of other signaling pathways in the regulation of CAR and PXR. The role of WNT/β-catenin signaling in P450 regulation has been studied using the natural WNT ligand WNT3a and siRNA-mediated depletion of β-catenin. Thomas et al. (Citation2015) showed that β-catenin is required for induction of downstream P450 genes by AhR, PXR and CAR, whereas it opposes induction by PPARα, while Chen et al. (Citation2018) performed gene knockdowns of CAR, PXR, hepatocyte nuclear factor 1α (HNF1α) and hepatocyte nuclear factor 4α (HNF4α) on P450 expression in HepaRGs. Knocking down PXR or CAR had little effect on the expression of other transcription factors but reduced the basal and induced expression of CYP2B6 and CYP3A4. Knocking down HNF1α or HNF4α upregulated AhR but decreased the expression of PXR, CAR, and many P450s, possibly via effects on PXR and CAR. Further studies are needed to resolve the mechanisms involved.

One potential complicating factor in the interpretation of induction studies using HepG2s and HepaRGs is the lack of information on key regulatory pathways in these cell lines. Without a complete description of the cell lines’ regulatory phenotypes, one cannot be sure whether the relatively low inducibility observed is due to the inability of the inducing agents to access receptors, failures in downstream signaling pathways, or flaws in the cells’ xenobiotic-metabolizing machinery (e.g. low levels of P450 oxidoreductase or essential cofactors).

Regulatory acceptance

The wide variability in cell culture, differentiation, and induction protocols reported across the HepG2 and HepaRG literature have, historically, made it very difficult to compare the results reported by different research groups, calling into question the utility of these cell lines for regulatory use. In addition, in vitro induction studies using PHHs, HepaRG and HepG2s commonly do not make direct comparisons with induction in vivo; when they do the results obtained often fail to reflect induction ratios observed in human volunteers. Berger et al. (Citation2016), for example, treated fifteen healthy volunteers with rifampicin (600 mg/day for 7 days) and calculated clearance ratios of probe drugs to assess the extent of P450 induction. They found that PHHs cultured in 3 D format best reflected in vivo induction; the results in HepaRGs resembled those in 3 D PHHs but correlated less well with induction in human volunteers in vivo.

The need for a consistent approach to in vitro induction studies has been recognized and various regulatory agencies have initiated programs aimed at the development of standardized protocols for the prediction of in vivo induction using PHHs and HepaRGs. The European Center for the Validation of Alternative Methods, in collaboration with the EU Reference Laboratories, has developed and validated protocols for induction studies in cryopreserved human PHHs and HepaRGs (Bernasconi et al. Citation2019). Comparing pharmaceuticals for which human in vivo induction data exist with BNF, PB, and rifampicin, they concluded that cryopreserved PHHs and HepaRGs respond appropriately to the three canonical inducing agents and most blinded test items when induction is assessed using a cocktail of phenacetin, bupropion, and midazolam and metabolites are quantified by LC/MS. The best outcome was for induction of CYP3A4, which was correctly predicted in every case except for artemisinin. The HepaRG method was subsequently transferred to the Organization for Economic Co-operation and Development (OECD) Test Guideline development program (https://tsar.jrc.ec.europa.eu/test-method/tm2009-14); however, the OECD decided against taking the PHH method forward (https://tsar.jrc.ec.europa.eu/test-method/tm2009-13).

A draft OECD Test Guideline, Determination of cytochrome P450 (CYP) enzyme activity induction using differentiated human hepatic cells was issued in October 2019 (OECD Citation2019). This defines a protocol for induction studies in human derived-metabolically competent hepatic test systems (e.g. cryopreserved differentiated HepaRGs) and highlights key considerations, including the use of DMSO at concentrations below 0.1% in assay medium to avoid interference with PXR (presumably after the completion of the differentiation process, since this usually requires the use of ∼2% DMSO). It recommends the use of enzyme activity measurements, rather than mRNA upregulation, as the parameter by which to quantify induction, noting that:

The measurement of functional CYP enzyme activity induction (i.e., catalytic activity) is considered more informative and relevant for chemical risk assessment than measurement of mRNA, since correlations between the CYP-selective activity and the specific CYP mRNA level are frequently poor or lacking.

This problem has been reported by users of in vitro induction assays in the pharmaceutical industry, who report that they sometimes observe increases in mRNA expression without corresponding upregulation of enzyme activity (Chu et al. Citation2009), particularly when a test item is both an inducer and an inhibitor. Even where induction is observed at both the mRNA and protein level, mRNA results tend to overestimate maximal induction (Emax) values by up to 10-fold, sometimes more, especially in the case of time-dependent inhibitors of CYP3A4, such as troleandomycin, verapamil, and ritonavir (McGinnity et al. Citation2009).

The test protocol, referring to Kanebratt and Andersson (Citation2008b), classifies a test chemical with ≥2-fold CYP enzyme activity induction compared to negative control as an in vitro positive inducer, resembling the detection limits of 1.6-fold against a water vehicle control and 1.7-fold against 0.1% DMSO determined by Kaneko et al. (Citation2009). It also specifies quality assurance criteria along with a panel of chemicals to be used to demonstrate technical proficiency before attempting to characterize induction by test compounds.

The US Federal Drug Administration (USFDA) will accept induction data obtained using PHHs or immortalized cell lines. No preference is expressed as to which cell line should be used; the only explicit mention of HepaRGs in this guidance is a citation of normalization methods for the prediction of in vitro CYP3A4 induction (Vermet et al. Citation2016); instead, the guidance focuses on the use of well-characterized individual batches of cryopreserved PHHs from at least three donors. In terms of the endpoints used, ‘Acceptable study endpoints include mRNA levels and/or enzyme activity levels using a probe substrate.’ A 2-fold change in mRNA expression is considered acceptable (subject to other criteria having been met, including justification of the test system used); however, no fold induction level is specified for cases where enzyme activity, rather than mRNA expression, is measured.

This guidance may be helpful in standardizing induction studies using HepaRGs; however, Kaneko et al. (Citation2009) note that they were unable to predict the inducing potential of PB, carbamazepine, sulfinpyrazone, or phenytoin using the methodology recommended in a previous (2006) version of the USFDA guidance.

Both the draft OECD Guideline and the USFDA guidance specify 2-fold induction as being indicative of potential drug-drug interaction liabilities; however, it is important to note that the informativeness of criteria, such as ‘fold induction over vehicle control’ or ‘% of positive control’ is dependent upon the basal expression level, the magnitude of the positive response and the variability of the results: 40% of a 5-fold positive control response would likely be within the background variability of the assay (Chu et al. Citation2009). It is also, of course, critical to relate any observations in vitro to likely exposure levels in vivo.

Evaluation

The HepG2 cell line has a primitive hepatocyte phenotype consistent with its hepatoblastoma origin (Knowles et al. Citation1980) and its proteome resembles that of human fetal hepatocytes rather than adult cells (Rowe et al. Citation2013). This is reflected in its expression of higher levels of CYP3A7 than CYP3A4/5 and its CYP1A profile, which favors the expression of CYP1A1 over that of CYP1A2 (Ulvestad et al. Citation2013). Consistent with its origin, it retains some AhR functionality and responds robustly to CYP1A inducing agents, but it is essentially refractory to PXR and CAR ligands. Thus, while HepG2s are recognized as being well-characterized and widely used in drug metabolism and toxicology studies, their low levels of CYP3A4 and lack of response to known CYP3A4 inducing agents limit their utility in regulatory studies, such as those aimed at the prediction of drug-drug interactions (Chu et al. Citation2009).

HepaRGs, when fully differentiated, bear some resemblance to adult human hepatocytes. The group which established them considers them to be representative of PHHs with the advantage of being reproducible over several passages, with functional activities which are maintained for several weeks at the confluence and can be modulated by manipulating the culture conditions (Anthérieu et al. Citation2010). Other investigators, however, have struggled to detect P450-dependent activities in these cells. The overall picture of their functionality is confusing and contradictory.

The need to culture HepaRGs in the presence of DMSO to maintain the differentiated phenotype raises concerns. The fact that the differentiation process requires a complex series of treatments and replatings, as well as the use of slightly different protocols by different investigators, has created huge study-to-study variability. Furthermore, DMSO is cytotoxic at the concentrations used; indeed, the original description of HepaRG cell differentiation notes that the majority of the cells die in the presence of 2% DMSO and it is only the small number of surviving cells that go on to differentiate (Gripon et al. Citation2002). The fact that DMSO alters gene expression in ways that have not been fully characterized also raises concerns; HepaRGs exhibit a biochemical profile that is markedly different from that of PHHs with respect to glucose homeostasis, lactate and urea production, galactose and sorbitol elimination, and albumin expression (Lübberstedt et al. Citation2011). Finally, DMSO is both an inducer of P450 expression and an inhibitor of P450 activity; indeed, it is recognized that the apparent poor responsiveness of HepaRGs to rifampicin induction may be a consequence of CYP3A4 expression having already been maximally induced by DMSO (Aninat et al. Citation2006; Lübberstedt et al. Citation2011). HepaRGs may therefore overestimate the contribution of CYP3A4 to hepatic drug metabolism while simultaneously underestimating induction by drugs that act via NRs, particularly PXR (Kanebratt and Andersson Citation2008a).

HepaRGs are widely considered to be a good tool for induction studies; however, they have numerous limitations, particularly wide batch-to-batch variability associated with the need to differentiate each batch before use (Kaneko et al. Citation2009), although the availability of pre-differentiated cryoHepaRGs goes some way toward overcoming this problem. The fact that the expression of P450 enzymes and NRs, as well as responsiveness to enzyme inducers, in this cell line varies so markedly in response to changes in media composition and culture conditions severely hampers its utility for induction studies, especially those conducted for regulatory purposes (Chu et al. Citation2009). Better transparency concerning the composition of the proprietary induction media and supplements which are promoted for use with this cell line would be of considerable assistance in overcoming this problem. The availability of DMSO-free differentiation media for HepaRGs (Wang et al. Citation2019) may help to resolve this issue if adopted and used according to a publicly available, standardized protocol. The use of HepaRG derivatives which can differentiate in the absence of DMSO (e.g. HepaRG-CAR) represents an alternative strategy, although having been further engineered such models are arguably even more unlike PHHs than are HepaRGs themselves.

Conclusions

Based on the available data neither HepG2 nor HepaRG is fully reflective of the drug-metabolizing capabilities of human hepatocytes in vitro, let alone in vivo. Indeed, no single in vitro model can accurately reflect the diverse array of outcomes required to mimic the hepatic function in vivo (Chu et al. Citation2009; Hurrell et al. Citation2018). Apart from the issues highlighted in this review, including the need for clear guidelines as to the necessary characterization of each batch of cells and consistent protocols for induction and activity measurements, it must be recognized that single-origin cell lines cannot represent other aspects of drug disposition including metabolism in the gastrointestinal tract, delivery to the hepatocyte via the portal circulation and factors affecting subsequent elimination. The best way to use these cell lines, therefore, is in combination with other models, such as mice humanized for CAR, PXR, and various P450s (Henderson et al. Citation2019), which allow induction capacity in vivo to be assessed and pharmacokinetic/pharmacodynamic relationships to be characterized in vivo and extrapolated to man. The value of humanized models for mechanistic studies has been recognized for more than a decade (Chu et al. Citation2009); they provide essential information about systemic effects, a dimension which cell culture models cannot offer.

Disclosure statement

CRW is not involved in the development of immortalized cell lines for use in drug metabolism studies but is involved in the use of humanized mouse models for predicting pathways of P450-mediated metabolism and DDIs in man. LAS is an independent Consultant in Investigative Toxicology but also holds a non-stipendiary appointment as an Associate of the School of Applied Sciences, Edinburgh Napier University. This review was prepared under a consultancy agreement with the University of Dundee.

References

  • Aninat C, Piton A, Glaise D, Le Charpentier T, Langouët S, Morel F, Guguen-Guillouzo C, Guillouzo A. 2006. Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells. Drug Metab Dispos. 34(1):75–83.
  • Anthérieu S, Chesné C, Li R, Camus S, Lahoz A, Picazo L, Turpeinen M, Tolonen A, Uusitalo J, Guguen-Guillouzo C, et al. 2010. Stable expression, activity, and inducibility of cytochromes P450 in differentiated HepaRG cells. Drug Metab Dispos. 38(3):516–525.
  • Berger B, Donzelli M, Maseneni S, Boess F, Roth A, Krähenbühl S, Haschke M. 2016. Comparison of liver cell models using the basel phenotyping cocktail. Front Pharmacol. 7:443.
  • Berger E, Vega N, Weiss-Gayet M, Géloën A. 2015. Gene network analysis of glucose linked signaling pathways and their role in human hepatocellular carcinoma cell growth and survival in HuH7 and HepG2 cell lines. Biomed Res Int. 2015:821761.
  • Bernasconi C, Pelkonen O, Andersson TB, Strickland J, Wilk-Zasadna I, Asturiol D, Cole T, Liska R, Worth A, Müller-Vieira U, et al. 2019. Validation of in vitro methods for human cytochrome P450 enzyme induction: outcome of a multi-laboratory study. Toxicol In Vitro. 60:212–228.
  • Bonn B, Svanberg P, Janefeldt A, Hultman I, Grime K. 2016. Determination of human hepatocyte intrinsic clearance for slowly metabolized compounds: comparison of a primary hepatocyte/stromal cell co-culture with plated primary hepatocytes and HepaRG. Drug Metab Dispos. 44(4):527–533.
  • Buccitelli C, Selbach M. 2020. mRNAs, proteins and the emerging principles of gene expression control. Nat Rev Genet. 21(10):630–644.
  • Chen L, Bao Y, Piekos SC, Zhu K, Zhang L, Zhong XB. 2018. A transcriptional regulatory network containing nuclear receptors and long noncoding RNAs controls basal and drug-induced expression of cytochrome P450s in HepaRG cells. Mol Pharmacol. 94(1):749–759.
  • Choi JM, Oh SJ, Lee JY, Jeon JS, Ryu CS, Kim YM, Lee K, Kim SK. 2015. Prediction of drug-induced liver injury in HepG2 cells cultured with human liver microsomes. Chem Res Toxicol. 28(5):872–885.
  • Chu V, Einolf HJ, Evers R, Kumar G, Moore D, Ripp S, Silva J, Sinha V, Sinz M, Skerjanec A. 2009. In vitro and in vivo induction of cytochrome P450: a survey of the current practices and recommendations: a pharmaceutical research and manufacturers of America perspective. Drug Metab Dispos. 37(7):1339–1354.
  • Dawson JR, Adams DJ, Wolf CR. 1985. Induction of drug metabolizing enzymes in human liver cell line HepG2. FEBS Lett. 183(2):219–222.
  • Dearfield KL, Jacobson-Kram D, Brown NA, Williams JR. 1983. Evaluation of a human hepatoma cell line as a target cell in genetic toxicology. Mutat Res. 108(1–3):437–449.
  • Easterbrook J, Lu C, Sakai Y, Li AP. 2001. Effects of organic solvents on the activities of cytochrome P450 isoforms, UDP-dependent glucuronyl transferase, and phenol sulfotransferase in human hepatocytes. Drug Metab Dispos. 29(2):141–144.
  • El-Sankary W, Plant NJ, Gibson GG, Moore DJ. 2000. Regulation of the CYP3A4 gene by hydrocortisone and xenobiotics: role of the glucocorticoid and pregnane X receptors. Drug Metab Dispos. 28(5):493–496.
  • Gerets HH, Tilmant K, Gerin B, Chanteux H, Depelchin BO, Dhalluin S, Atienzar FA. 2012. Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biol Toxicol. 28(2):69–87.
  • Gripon P, Rumin S, Urban S, Le Seyec J, Glaise D, Cannie I, Guyomard C, Lucas J, Trepo C, Guguen-Guillouzo C. 2002. Infection of a human hepatoma cell line by hepatitis B virus. Proc Natl Acad Sci USA. 99(24):15655–15660.
  • Hammer H, Schmidt F, Marx-Stoelting P, Pötz O, Braeuning A. 2021. Cross-species analysis of hepatic cytochrome P450 and transport protein expression. Arch Toxicol. 95(1):117–133.
  • Harmsen S, Koster AS, Beijnen JH, Schellens JH, Meijerman I. 2008. Comparison of two immortalized human cell lines to study nuclear receptor-mediated CYP3A4 induction. Drug Metab Dispos. 36(6):1166–1171.
  • Henderson CJ, Kapelyukh Y, Scheer N, Rode A, McLaren AW, MacLeod AK, Lin D, Wright J, Stanley LA, Wolf CR. 2019. An extensively humanized mouse model to predict pathways of drug disposition and drug/drug interactions, and to facilitate design of clinical trials. Drug Metab Dispos. 47(6):601–615.
  • Hurrell T, Segeritz CP, Vallier L, Lilley KS, Cromarty AD. 2018. Proteomic comparison of various hepatic cell cultures for preclinical safety pharmacology. Toxicol Sci. 164(1):229–239.
  • Jackson JP, Li L, Chamberlain ED, Wang H, Ferguson SS. 2016. Contextualizing hepatocyte functionality of cryopreserved HepaRG cell cultures. Drug Metab Dispos. 44(9):1463–1479.
  • Jossé R, Aninat C, Glaise D, Dumont J, Fessard V, Morel F, Poul JM, Guguen-Guillouzo C, Guillouzo A. 2008. Long-term functional stability of human HepaRG hepatocytes and use for chronic toxicity and genotoxicity studies. Drug Metab Dispos. 36(6):1111–1118.
  • Kanebratt KP, Andersson TB. 2008a. Evaluation of HepaRG cells as an in vitro model for human drug metabolism studies. Drug Metab Dispos. 36(7):1444–1452.
  • Kanebratt KP, Andersson TB. 2008b. HepaRG cells as an in vitro model for evaluation of cytochrome P450 induction in humans. Drug Metab Dispos. 36(1):137–145.
  • Kaneko A, Kato M, Sekiguchi N, Mitsui T, Takeda K, Aso Y. 2009. In vitro model for the prediction of clinical CYP3A4 induction using HepaRG cells. Xenobiotica. 39(11):803–810.
  • Kelly JH, Sussman NL. 2000. A fluorescent cell-based assay for cytochrome P-450 isozyme 1A2 induction and inhibition. J Biomol Screen. 5(4):249–254.
  • Khoo KC, Mendels J, Rothbart M, Garland WA, Colburn WA, Min BH, Lucek R, Carbone JJ, Boxenbaum HG, Kaplan SA. 1980. Influence of phenytoin and phenobarbital on the disposition of a single oral dose of clonazepam. Clin Pharmacol Ther. 28(3):368–375.
  • Knowles BB, Howe CC, Aden DP. 1980. Human hepatocellular carcinoma cell lines secrete the major plasma proteins and hepatitis B surface antigen. Science. 209(4455):497–499.
  • Koutsounas I, Theocharis S, Patsouris E, Giaginis C. 2013. Pregnane X receptor (PXR) at the crossroads of human metabolism and disease. Curr Drug Metab. 14(3):341–350.
  • Kratochwil NA, Meille C, Fowler S, Klammers F, Ekiciler A, Molitor B, Simon S, Walter I, McGinnis C, Walther J, et al. 2017. Metabolic profiling of human long-term liver models and hepatic clearance predictions from in vitro data using nonlinear mixed-effects modeling. Aaps J. 19(2):534–550.
  • Kvist AJ, Kanebratt KP, Walentinsson A, Palmgren H, O'Hara M, Björkbom A, Andersson LC, Ahlqvist M, Andersson TB. 2018. Critical differences in drug metabolic properties of human hepatic cellular models, including primary human hepatocytes, stem cell derived hepatocytes, and hepatoma cell lines. Biochem Pharmacol. 155:124–140.
  • Lübberstedt M, Müller-Vieira U, Mayer M, Biemel KM, Knöspel F, Knobeloch D, Nüssler AK, Gerlach JC, Zeilinger K. 2011. HepaRG human hepatic cell line utility as a surrogate for primary human hepatocytes in drug metabolism assessment in vitro. J Pharmacol Toxicol Methods. 63(1):59–68.
  • McGinnity DF, Zhang G, Kenny JR, Hamilton GA, Otmani S, Stams KR, Haney S, Brassil P, Stresser DM, Riley RJ. 2009. Evaluation of multiple in vitro systems for assessment of CYP3A4 induction in drug discovery: human hepatocytes, pregnane X receptor reporter gene, and Fa2N-4 and HepaRG cells. Drug Metab Dispos. 37(6):1259–1268.
  • McMahon M, Ding S, Jimenez LA, Terranova R, Gerard MA, Vitobello A, Moggs J, Henderson CJ, Wolf CR. 2019. Constitutive androstane receptor 1 is constitutively bound to chromatin and ‘primed’ for transactivation in hepatocytes. Mol Pharmacol. 95(1):97–105.
  • Negishi M, Kobayashi K, Sakuma T, Sueyoshi T. 2020. Nuclear receptor phosphorylation in xenobiotic signal transduction. J Biol Chem. 295(45):15210–15225.
  • Nelson LJ, Morgan K, Treskes P, Samuel K, Henderson CJ, LeBled C, Homer N, Grant MH, Hayes PC, Plevris JN. 2017. Human hepatic HepaRG cells maintain an organotypic phenotype with high intrinsic CYP450 activity/metabolism and significantly outperform standard HepG2/C3A cells for pharmaceutical and therapeutic applications. Basic Clin Pharmacol Toxicol. 120(1):30–37.
  • Nofziger C, Turner AJ, Sangkuhl K, Whirl-Carrillo M, Agúndez JAG, Black JL, Dunnenberger HM, Ruano G, Kennedy MA, Phillips MS, et al. 2020. PharmVar GeneFocus: CYP2D6. Clin Pharmacol Ther. 107(1):154–170.
  • OECD. 2019. Determination of Cytochrome P450 (CYP) enzyme activity induction using differentiated human hepatic cells. [accessed 2021 Aug 2]. https://www.oecd.org/chemicalsafety/testing/Draft_TG_CYP_induction_for_2nd_WNT_review.pdf
  • Parent R, Marion MJ, Furio L, Trépo C, Petit MA. 2004. Origin and characterization of a human bipotent liver progenitor cell line. Gastroenterology. 126(4):1147–1156.
  • Preiss LC, Liu R, Hewitt P, Thompson D, Georgi K, Badolo L, Lauschke VM, Petersson C. 2021. Deconvolution of cytochrome P450 induction mechanisms in HepaRG nuclear hormone receptor knockout cells. Drug Metab Dispos. 49(8):668–678.
  • Ramaiahgari SC, Waidyanatha S, Dixon D, DeVito MJ, Paules RS, Ferguson SS. 2017. From the cover: three-dimensional (3D) HepaRG spheroid model with physiologically relevant xenobiotic metabolism competence and hepatocyte functionality for liver toxicity screening. Toxicol Sci. 159(1):124–136.
  • Rowe C, Gerrard DT, Jenkins R, Berry A, Durkin K, Sundstrom L, Goldring CE, Park BK, Kitteringham NR, Hanley KP, et al. 2013. Proteome-wide analyses of human hepatocytes during differentiation and dedifferentiation. Hepatology. 58(2):799–809.
  • Sangkuhl K, Claudio-Campos K, Cavallari LH, Agundez JAG, Whirl-Carrillo M, Duconge J, Del Tredici AL, Wadelius M, Rodrigues Botton M, Woodahl EL, et al. 2021. PharmVar GeneFocus: CYP2C9. Clin Pharmacol Ther. 110(3):662–676.
  • Scheer N, Ross J, Rode A, Zevnik B, Niehaves S, Faust N, Wolf CR. 2008. A novel panel of mouse models to evaluate the role of human pregnane X receptor and constitutive androstane receptor in drug response. J Clin Invest. 118(9):3228–3239.
  • Seo JE, Tryndyak V, Wu Q, Dreval K, Pogribny I, Bryant M, Zhou T, Robison TW, Mei N, Guo X. 2019. Quantitative comparison of in vitro genotoxicity between metabolically competent HepaRG cells and HepG2 cells using the high-throughput high-content CometChip assay. Arch Toxicol. 93(5):1433–1448.
  • Seo JE, Wu Q, Bryant M, Ren L, Shi Q, Robison TW, Mei N, Manjanatha MG, Guo X. 2020. Performance of high-throughput CometChip assay using primary human hepatocytes: a comparison of DNA damage responses with in vitro human hepatoma cell lines. Arch Toxicol. 94(6):2207–2224.
  • Shi J, Wang X, Lyu L, Jiang H, Zhu HJ. 2018. Comparison of protein expression between human livers and the hepatic cell lines HepG2, Hep3B, and Huh7 using SWATH and MRM-HR proteomics: Focusing on drug-metabolizing enzymes. Drug Metab Pharmacokinet. 33(2):133–140.
  • Sison-Young RL, Mitsa D, Jenkins RE, Mottram D, Alexandre E, Richert L, Aerts H, Weaver RJ, Jones RP, Johann E, et al. 2015. Comparative proteomic characterization of 4 human liver-derived single cell culture models reveals significant variation in the capacity for drug disposition, bioactivation, and detoxication. Toxicol Sci. 147(2):412–424.
  • Stanley LA, Horsburgh BC, Ross J, Scheer N, Wolf CR. 2006. PXR and CAR: nuclear receptors which play a pivotal role in drug disposition and chemical toxicity. Drug Metab Rev. 38(3):515–597.
  • Tascher G, Burban A, Camus S, Plumel M, Chanon S, Le Guevel R, Shevchenko V, Van Dorsselaer A, Lefai E, Guguen-Guillouzo C, et al. 2019. In-depth proteome analysis highlights HepaRG cells as a versatile cell system surrogate for primary human hepatocytes. Cells. 8(2):192.
  • Thomas M, Bayha C, Vetter S, Hofmann U, Schwarz M, Zanger UM, Braeuning A. 2015. Activating and inhibitory functions of WNT/β-catenin in the induction of cytochromes P450 by nuclear receptors in HepaRG cells. Mol Pharmacol. 87(6):1013–1020.
  • Tolosa L, Gómez-Lechón MJ, López S, Guzmán C, Castell JV, Donato MT, Jover R. 2016. Human upcyte hepatocytes: characterization of the hepatic phenotype and evaluation for acute and long-term hepatotoxicity routine testing. Toxicol Sci. 152(1):214–229.
  • Turpeinen M, Tolonen A, Chesne C, Guillouzo A, Uusitalo J, Pelkonen O. 2009. Functional expression, inhibition and induction of CYP enzymes in HepaRG cells. Toxicol In Vitro. 23(4):748–753.
  • Ulvestad M, Nordell P, Asplund A, Rehnström M, Jacobsson S, Holmgren G, Davidson L, Brolén G, Edsbagge J, Björquist P, et al. 2013. Drug metabolizing enzyme and transporter protein profiles of hepatocytes derived from human embryonic and induced pluripotent stem cells. Biochem Pharmacol. 86(5):691–702.
  • van der Mark VA, Rudi de Waart D, Shevchenko V, Elferink RP, Chamuleau RA, Hoekstra R. 2017. Stable overexpression of the constitutive androstane receptor reduces the requirement for culture with dimethyl sulfoxide for high drug metabolism in HepaRG cells. Drug Metab Dispos. 45(1):56–67.
  • Vermet H, Raoust N, Ngo R, Esserméant L, Klieber S, Fabre G, Boulenc X. 2016. Evaluation of normalization methods to predict CYP3A4 induction in six fully characterized cryopreserved human hepatocyte preparations and HepaRG cells. Drug Metab Dispos. 44(1):50–60.
  • Wang Z, Luo X, Anene-Nzelu C, Yu Y, Hong X, Singh NH, Xia L, Liu S, Yu H. 2015. HepaRG culture in tethered spheroids as an in vitro three-dimensional model for drug safety screening. J Appl Toxicol. 35(8):909–917.
  • Wang ZY, Li WJ, Li QG, Jing HS, Yuan TJ, Fu GB, Tang D, Zhang HD, Yan HX, Zhai B. 2019. A DMSO-free hepatocyte maturation medium accelerates hepatic differentiation of HepaRG cells in vitro. Biomed Pharmacother. 116:109010.
  • Weaver RJ, Betts C, Blomme EAG, Gerets HHJ, Gjervig Jensen K, Hewitt PG, Juhila S, Labbe G, Liguori MJ, Mesens N, et al. 2017. Test systems in drug discovery for hazard identification and risk assessment of human drug-induced liver injury. Expert Opin Drug Metab Toxicol. 13(7):767–782.
  • Weaver RJ, Blomme EA, Chadwick AE, Copple IM, Gerets HHJ, Goldring CE, Guillouzo A, Hewitt PG, Ingelman-Sundberg M, Jensen KG, et al. 2020. Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat Rev Drug Discov. 19(2):131–148.
  • Williamson B, Lorbeer M, Mitchell MD, Brayman TG, Riley RJ. 2016. Evaluation of a novel PXR-knockout in HepaRG™ cells. Pharmacol Res Perspect. 4(5):e00264.
  • Wiśniewski JR, Vildhede A, Norén A, Artursson P. 2016. In-depth quantitative analysis and comparison of the human hepatocyte and hepatoma cell line HepG2 proteomes. J Proteomics. 136:234–247.
  • Wong N, Lai P, Pang E, Leung TW, Lau JW, Johnson PJ. 2000. A comprehensive karyotypic study on human hepatocellular carcinoma by spectral karyotyping. Hepatology. 32(5):1060–1068.
  • Xie Y, Ke S, Chen J, Ouyang N, Tian Y. 2020. Epigenetic sensitization of pregnane X receptor-regulated gene expression by dimethyl sulfoxide. Toxicol Lett. 321:131–137.
  • Yokoyama Y, Sasaki Y, Terasaki N, Kawataki T, Takekawa K, Iwase Y, Shimizu T, Sanoh S, Ohta S. 2018. Comparison of drug metabolism and its related hepatotoxic effects in HepaRG, cryopreserved human hepatocytes, and HepG2 cell cultures. Biol Pharm Bull. 41(5):722–732.
  • Zanger UM, Schwab M. 2013. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 138(1):103–141.
  • Zhou B, Ho SS, Greer SU, Spies N, Bell JM, Zhang X, Zhu X, Arthur JG, Byeon S, Pattni R, et al. 2019. Haplotype-resolved and integrated genome analysis of the cancer cell line HepG2. Nucleic Acids Res. 47(8):3846–3861.