Elsevier

Toxicology in Vitro

Volume 30, Issue 1, Part A, 25 December 2015, Pages 217-224
Toxicology in Vitro

Biokinetics in repeated-dosing in vitro drug toxicity studies

https://doi.org/10.1016/j.tiv.2015.09.005Get rights and content

Highlights

  • This paper reviews work done within WP3 of the EU FP7 Predict-IV project.

  • RPTEC/TERT1, HepaRG, primary hepatocytes and brain cultures were considered.

  • Medium dosed with drugs was refreshed daily or every other day for 14 days.

  • Drug concentrations over time were measured and modelled in various system phases.

  • Drugs differentially accumulate in cells in repeatedly-dosed in vitro assays.

Abstract

The aim of the EU FP7 Predict-IV project was to improve the predictivity of in vitro assays for unwanted effects of drugs after repeated dosing. The project assessed the added benefit of integrating long-lived in vitro organotypic cell systems with ‘omics’ technologies and in silico modelling, including systems biology and pharmacokinetic assessments. RPTEC/TERT1 kidney cells, primary rat and human hepatocytes, HepaRG liver cells and 2D and 3D primary brain cultures were dosed daily or every other day for 14 days to a selection of drugs varying in their mechanism of pharmacological action. Since concentration–effect relationships not only depend on the activity of the drug or the sensitivity of the target, but also on the distribution of compounds in the in vitro system, the concentration of a selection of drugs in cells, microtitre plate plastic and medium was measured over time. Results, reviewed in this paper, indicate that lipophilic drugs bind significantly to plastic labware. A few drugs, including less lipophilic drugs, bind to cell-attachment matrices. Chemicals that reach high concentrations in cells, including cyclosporin A and amiodarone, significantly accumulate over time after repeated dosing, partly explaining their increased toxicity after repeated dosing, compared to a single dose.

Introduction

Many derivatives of a lead compound exhibiting a desired pharmacological effect are synthesised early on in the development of a new pharmaceutical entity in order to identify the ones with the most optimal pharmacological response. In order to assess the safety of these lead compounds before the ‘first dose in man’, toxicity testing of a large number of chemicals on animals is necessary and a costly endeavour (Sasseville et al., 2004). This testing represents one of the major bottlenecks in drug development as toxicity testing in preclinical studies is time consuming and requires large numbers of animals and considerable amounts of test compound. In addition, the high costs of toxicity testing are exacerbated by the high drug attrition rate, where 23% of registered drugs are retracted due to adverse reactions not predicted in animal models (Kola, 2008). These adverse reactions are often idiosyncratic and occur after repeated dosing. Major reasons for the suboptimal correlation between animal and human toxicity are the inter- and intra-species differences in pharmacokinetics (Park et al., 2011).

In light of both ethical and financial costs associated with drug safety testing on animals, human cell-based in vitro assays are increasingly used to screen drug candidates for human-relevant pharmacokinetic properties and molecular mechanisms of toxicity prior to pre-clinical testing in animals. However, the move from using in vitro assays for hazard identification, i.e. the mere potential of a chemical to cause an effect, to hazard characterization in drug development, i.e. dose–response assessment, is still in its early stage of development. It is generally accepted that no single stand-alone in vitro test sufficiently replaces an animal-based toxicity test and thus an integrated strategy is required. Such strategy calls for a battery of in vitro assays employing long-lived, highly functional organotypic cell cultures and a mechanistic understanding of the molecular events leading to adverse health effects (Adler et al., 2011). For such in vitro test battery to be used in a risk assessment procedure, a point of departure needs to be derived from the set of dose–response relationships obtained from these assays and translated into a toxicologically equivalent dose in humans. Indeed, the pharmacokinetics (i.e. the absorption, distribution, metabolism and excretion from a body) of a drug determines the concentration over time of the drug (or its toxicologically relevant metabolite) at the target site, which strongly dictates the drug's toxicity. These processes need to be integrated into a meaningful in vitro-based drug safety testing strategy (Adler et al., 2011).

To improve the predictivity of in vitro systems for unwanted effects of drugs after repeated dosing, the aim of the EU 7th Framework Project, Predict-IV, was to develop such a testing strategy integrating in vitro systems with knowledge of cell biology, mechanistic toxicology and in silico (pharmacokinetic) modelling. The project focussed its efforts on developing testing strategies by using in vitro assays with cells of human origin (whenever possible) and representing target organs most frequently affected by poorly predicted drug toxicity, namely the liver, kidney and central nervous system. It opted for using primary human hepatocytes, primary rat hepatocytes and the human hepatoma cell line HepaRG as its main liver models (Mueller et al., 2015), 2D mouse and 3D rat primary brain cell models with an in vitro blood brain barrier (BBB) model to predict neurotoxicity (Culot et al., 2008, Culot et al., 2013, Schultz et al., 2015), and the human renal proximal tubule cell line RPTEC/TERT1 (Wieser et al., 2008, Aschauer et al., 2015b) as its model of choice for predicting nephrotoxicity. Culture conditions were adapted to maintain highly differentiated organotypic cells in culture for 14 days, during which the cells were exposed daily to a selection of 27 drugs varying in their mechanism of pharmacological action and known to cause hepatotoxicity, nephrotoxicity and/or neurotoxicity after repeated use. For a holistic, mechanistic approach, in depth characterization of molecular perturbations induced by the drugs was performed by integrating a suite of ‘omics’ technologies (e.g. Wilmes et al., 2013). Moreover, exposure conditions and changes within the assays over the 14-day exposure period were monitored and modelled (e.g. Pomponio et al., 2015a, Pomponio et al., 2015b, Truisi et al., 2015). Dose response analyses and physiologically based pharmacokinetic (PBPK) models were developed to relate daily oral exposure to in vitro derived points of departures (Hamon et al., 2015).

Section snippets

Role of in vitro biokinetics in quantitative in vitro–in vivo extrapolation (QIVIVE) studies

Predict-IV uniquely devoted a separate work package (WP3 ‘Non animal-based models for in vitro kinetics and human kinetic prediction’) to propose and apply a step-wise strategy to measure and model cell exposure levels over time of a selected number of drugs in the developed in vitro assays. The aim was to assess whether and how knowledge of the kinetics of drugs in in vitro assays helps to explain the variations in observed effects between drugs, cell types and assay setup, and – in so doing –

Chemical stability

Chemical stability in solution determines the concentration in cells and subsequently its potential to perturb molecular pathways in vitro. The concentration in stock solutions and exposure medium of drugs listed in Table 1 were measured over time. Whereas most drugs were chemically stable in exposure medium as well as in the vehicles used to prepare stock solutions, i.e. distilled water, methanol and DMSO, adefovir dipivoxil hydrolysed significantly in exposure medium of RTPEC/TERT1 cells at 37

Expression and activity of drug transporters and biotransformation enzymes

To identify specific mechanisms of toxicity and new toxicity pathways, it is important to distinguish between an increase in cytotoxic potency of a drug in vitro that is due to higher intracellular concentrations of the drug (i.e. due to differences in kinetics) and an intrinsic sensitivity of a cell type to a drug (i.e. the drug's ability to activate selective toxicity pathways). If a significant fraction of a drug is taken up by cells, repeated dosing of the cells, as done in the Predict-IV

Conclusions: a strategy for future in vitro testing

The aim of the EU FP7 Predict-IV project was to improve the predictivity of in vitro assays for unwanted effects of drugs after repeated dosing. The project assessed the added benefit of integrating long-lived in vitro cell systems, representing the liver, the kidney and the central nervous system, with ‘omics’ technologies and in silico modelling, including systems biology and pharmacokinetic assessments. WP3 studied the distribution over time (i.e. kinetics) of a selection of drugs, varying

Acknowledgements

The authors declare no conflicts of interest. This work was supported by the EU FP 7 Predict-IV programme (grant 202222). The authors are grateful to the many PhD students and scientists performing the work cited in this overview paper. The authors would also like to acknowledge the Doerenkamp Zbinden Foundation for financially supporting N.I. Kramer and B.J. Blaauboer at Utrecht University.

References (62)

  • A. Fabulas-Da Costa et al.

    In vitro blood–brain barrier model adapted to repeated-dose toxicological screening

    Toxicol. In Vitro

    (2013)
  • F.A. Groothuis et al.

    Dose metric considerations in in vitro assays to improve quantitative in vitroin vivo dose extrapolations

    Toxicology

    (2015)
  • M. Gülden et al.

    Factors influencing nominal effective concentrations of chemical compounds in vitro: cell concentration

    Toxicol. In Vitro

    (2001)
  • M. Gülden et al.

    In vitro toxicity testing with microplate cell cultures: impact of cell binding

    Toxicology

    (2015)
  • M. Gülden et al.

    Impact of bioavailability on the correlation between in vitro cytotoxic and in vivo acute fish toxic concentrations of chemicals

    Aquat. Toxicol.

    (2005)
  • M. Gülden et al.

    In vitroin vivo extrapolation: estimation of human serum concentrations of chemicals equivalent to cytotoxic concentrations in vitro

    Toxicology

    (2003)
  • J. Hamon et al.

    Quantitative in vitro to in vivo extrapolation of tissues toxicity

    Toxicol. In Vitro

    (2015)
  • M.B. Heringa et al.

    Measurement of free concentrations using negligible depletion-solid phase microextraction (nd-SPME)

    TrAC, Trends Anal. Chem.

    (2003)
  • M. Lübberstedt et al.

    HepaRG human hepatic cell line utility as a surrogate for primary human hepatocytes in drug metabolism assessment in vitro

    J. Pharmacol. Toxicol. Methods

    (2011)
  • S.O. Mueller et al.

    Drug biokinetics and toxicity assessment in rat and human primary hepatocytes and HepaRG cells within the EU-funded Predict-IV project

    Toxicol. In Vitro

    (2015)
  • G. Pomponio et al.

    In vitro kinetics of amiodarone and its major metabolite in two human liver cell models after acute and repeated treatments

    Toxicol. In Vitro

    (2015)
  • G. Pomponio et al.

    Amiodarone biokinetics, the formation of its major oxidative metabolite and neurotoxicity after acute and repeated exposure of brain cell cultures

    Toxicol. In Vitro

    (2015)
  • J. Riedl et al.

    Physicochemical substance properties as indicators for unreliable exposure in microplate-based bioassays

    Chemosphere

    (2007)
  • V.G. Sasseville et al.

    Testing paradigm for prediction of development-limiting barriers and human drug toxicity

    Chem. Biol. Interact.

    (2004)
  • C.C. Savary et al.

    Transcriptomic analysis of untreated and drug-treated differentiated HepaRG cells over a 2-week period

    Toxicol. In Vitro

    (2015)
  • K. Schirmer et al.

    Methodology for demonstrating and measuring the photocytotoxicity of fluoranthene to fish cells in culture

    Toxicol. In Vitro

    (1997)
  • L. Schultz et al.

    Evaluation of drug-induced neurotoxicity based on metabolomics, proteomics and electrical activity measurements in complementary CNS in vitro models

    Toxicol. In Vitro

    (2015)
  • H. Seibert et al.

    Factors influencing nominal effective concentrations of chemical compounds in vitro: medium protein concentration

    Toxicol. In Vitro

    (2002)
  • V. Tirelli et al.

    Effects of the pesticide chlorpyrifos on an in vitro model of intestinal barrier

    Toxicol. In Vitro

    (2007)
  • G.L. Truisi et al.

    Understanding the biokinetics of ibuprofen after single and repeated treatments in rat and human in vitro liver cell systems

    Toxicol. Lett.

    (2015)
  • S. Vichi et al.

    Cell type-specific expression and localization of cytochrome P450 isoforms in tridimensional aggregating rat brain cell cultures

    Toxicol. In Vitro

    (2015)
  • Cited by (71)

    View all citing articles on Scopus
    View full text