Characterizing uncertainty and population variability in the toxicokinetics of trichloroethylene and metabolites in mice, rats, and humans using an updated database, physiologically based pharmacokinetic (PBPK) model, and Bayesian approach

https://doi.org/10.1016/j.taap.2009.07.032Get rights and content

Abstract

We have developed a comprehensive, Bayesian, PBPK model-based analysis of the population toxicokinetics of trichloroethylene (TCE) and its metabolites in mice, rats, and humans, considering a wider range of physiological, chemical, in vitro, and in vivo data than any previously published analysis of TCE. The toxicokinetics of the “population average,” its population variability, and their uncertainties are characterized in an approach that strives to be maximally transparent and objective. Estimates of experimental variability and uncertainty were also included in this analysis. The experimental database was expanded to include virtually all available in vivo toxicokinetic data, which permitted, in rats and humans, the specification of separate datasets for model calibration and evaluation. The total combination of these approaches and PBPK analysis provides substantial support for the model predictions. In addition, we feel confident that the approach employed also yields an accurate characterization of the uncertainty in metabolic pathways for which available data were sparse or relatively indirect, such as GSH conjugation and respiratory tract metabolism. Key conclusions from the model predictions include the following: (1) as expected, TCE is substantially metabolized, primarily by oxidation at doses below saturation; (2) GSH conjugation and subsequent bioactivation in humans appear to be 10- to 100-fold greater than previously estimated; and (3) mice had the greatest rate of respiratory tract oxidative metabolism as compared to rats and humans. In a situation such as TCE in which there is large database of studies coupled with complex toxicokinetics, the Bayesian approach provides a systematic method of simultaneously estimating model parameters and characterizing their uncertainty and variability. However, care needs to be taken in its implementation to ensure biological consistency, transparency, and objectivity.

Introduction

Trichloroethylene (TCE) is a volatile organic solvent that has had widespread commercial and industrial use, particularly for industrial vapor degreasing of metal parts and as a chemical intermediate. TCE is also a common environmental contaminant at hazardous waste sites, in groundwater, and in ambient and indoor air. Because it is a dense nonaqueous-phase liquid, TCE is particularly difficult to remediate once it has entered groundwater [ATSDR, 1997, ATSDR, 2003].

Understanding TCE toxicokinetics is critical to both the qualitative and quantitative assessment of human health risks from environmental exposures and continues to be the subject of active research (Boyes et al., 2005, Chiu et al., 2007). Understanding TCE metabolism is especially important toxicologically because specific metabolites or metabolic pathways are associated with a number of endpoints of observed toxicity. In mice, TCE induces pulmonary toxicity after acute exposure (e.g., Forkert et al., 1985, Green et al., 1997, Odum et al., 1992) and lung tumors after chronic exposure (Fukuda et al., 1983; Maltoni et al., 1996), with available evidence suggesting that these effects are mediated by pulmonary oxidative metabolism (Forkert et al., 2005, Forkert et al., 2006). TCE and its oxidative metabolites trichloroacetic acid (TCA) and dichloroacetic acid (DCA) cause a number of similar effects in the liver of laboratory animals, including hepatomegaly in multiple rodent species, and hepatocarcinogenicity in multiple strains and sexes of mice (Bull, 2000). Moreover, the hepatomegalic effects in mice are more consistently associated with oxidative metabolism than with TCE itself (Buben and O'Flaherty, 1985, Evans et al., 2009). In the kidney, TCE causes tubular toxicity in mice and rats and is associated with small increases in the incidences of kidney tumors reported in multiple strains of TCE-exposed rats (Maltoni et al., 1986, National Cancer Institute NCI), (1976, National Toxicology Program NTP), (1988, National Toxicology Program NTP), (1990). These effects are thought to be mediated by products of glutathione (GSH) conjugation (Lash et al., 2000b, Lash et al., 2001). Human epidemiologic data also suggest that the kidney and liver are targets of TCE non-cancer toxicity (NRC, 2006). For carcinogenicity, the epidemiologic evidence is strongest for kidney cancer (NRC, 2006), with several well-designed and sufficiently powered studies reporting statistically significant increases in renal cell carcinoma incidence in groups occupationally exposed to TCE (e.g., Brüning et al., 2003, Charbotel et al., 2006, Zhao et al., 2005, Raaschou-Nielsen et al., 2003).

A simplified metabolism scheme for TCE is shown in Fig. 1. Briefly, as reviewed by Lash et al., 2000a, Chiu et al., 2006, metabolism of TCE occurs through two main irreversible pathways: oxidation via the microsomal mixed-function oxidase system (i.e., cytochrome P450s) and conjugation with GSH by glutathione S-transferases. For TCE oxidation, the P450 isoform CYP2E1 is thought to be most important in vivo. TCE is first oxidized to several unstable intermediate products (metabolites 2, 3, and 5), some of which are quickly transformed to trichloroethanol (TCOH-metabolite 6) (a reversible reaction) and trichloroacetic acid (TCA-metabolite 7). TCOH is glucuronidated to form TCOH-glucuronide (TCOG) (metabolite 8), which undergoes enterohepatic recirculation (excretion in bile with regeneration and reabsorption of TCOH from the gut). Both TCA and TCOG are excreted in urine, but other metabolism of TCA and TCOH has not been well characterized, although DCA (metabolite 9) has been hypothesized to be among the metabolism products (Lash et al., 2000a). TCE-oxide (metabolite 3) may also form DCA, among other species (Cai and Guengerich 1999). With respect to the conjugative pathway, dichlorovinyl glutathione (DCVG) (metabolite 4) is further processed to form the cysteine conjugate S-dichlorovinyl-l-cysteine (DCVC) (metabolite 10), which can undergo either bioactivation by beta-lyase or flavin-containing monooxygenases (FMO) to reactive species (Anders et al., 1988, Krause et al., 2003, Lash et al., 2003) or (reversible) N-acetylation to the mercapturate N-acetyl dichlorovinyl cysteine (NAcDCVC) (metabolite 11) excreted in urine or sulfoxidated by CYP3A to reactive species (Bernauer et al., 1996, Birner et al., 1993, Werner et al., 1995).

TCE has an extensive number of both in vivo pharmacokinetic and PBPK modeling studies in mice, rats, and humans (see Chiu et al. 2006, and Supplementary Material therein, for a review). Hack et al. (2006) represents the most recent published PBPK model attempting to integrate this vast database, using the Bayesian population approach first applied to TCE by Bois, 2000a, Bois, 2000b). A key question, then, is why should we develop yet another PBPK model for TCE and its metabolites? Indeed, our original intention was not to develop a completely revised model but rather to perform a minor “update” to the Hack et al (2006) effort. However, in the process of conducting a detailed evaluation of the Hack et al. (2006) model, the number of issues that required adjustment grew beyond what could reasonably be considered an application of the original model. The main conclusions of this evaluation, and their implications for PBPK model development, are summarized in Supplementary Materials (Table S-1). A deterministic analysis motivated the updated respiratory tract model and is reported separately (Evans et al., 2009).

In this article, we attempt to address these issues in conducting a new Bayesian, PBPK model-based, population toxicokinetic analysis of the currently available database of in vitro and in vivo toxicokinetic data in mice, rats, and humans. This analysis includes a much larger database of studies than has ever been published in a Bayesian PBPK analysis. Particular attention is paid to several important issues: (i) a fuller accounting of biologically plausible sources of uncertainty and variability; (ii) a clear separation between data used for developing prior estimates for model parameters and data used to update these estimates and generate posterior distributions, to avoid using the same data “twice”; (iii) explicit delineation of the type of variability being characterized in the population analysis, i.e., interstudy for mice and rats versus interindividual for humans; and (iv) a systematic evaluation of model convergence, posterior parameter estimates, and comparisons of model predictions with data. The resulting model with its parameter distributions is then used to characterize the uncertainty and variability in a number of important toxicokinetic processes, and can subsequently be used in probabilistic dose–response analyses (in preparation).

Section snippets

Updated PBPK model structure

The updated TCE PBPK model is illustrated in Fig. 2, with the changes from the Hack et al. (2006) model, and their justification, described in Table 1. The model reported here adds compartments for GSH conjugation metabolites for the rat and human to the model previously reported by Evans et al. (2009) in mice. In brief, the TCE sub-model was augmented/enhanced by the addition of kidney and venous blood compartments, and an updated respiratory tract model that included both metabolism and the

Convergence

The mouse model had the most rapid reduction in potential scale reduction factors. As reported in Evans et al. (2009), initially, four chains of 42,500 iterations each were run, with the first 12,500 discarded as “burn-in” iterations, i.e., iterations for which the simulation had not yet converged. The initial decision for determining “burn-in” was determined by visual inspection. At this point, all the population parameters except for the VMax for DCVG formation had R < 1.2, with only the first

Discussion

While its outputs are complex to analyze and synthesize, the hierarchical Bayesian analysis reported in this paper provides an opportunity to improve quantification of uncertainty and variability in TCE toxicokinetics. Specifically for TCE, this analysis substantially informs four of the major areas of pharmacokinetic uncertainty previously identified in numerous reports (reviewed in Chiu et al. 2006): GSH conjugation pathway, respiratory tract metabolism, alternative pathways of TCE oxidation

Disclaimer

This article has been reviewed by the US Environmental Protection Agency and approved for publication. The views expressed in this manuscript are those of the authors and do not necessarily reflect the views or policies of the US Environmental Protection Agency.

Conflict of Interest statement

The authors declare that there are no conflicts of interest.

Acknowledgments

The authors would like to thank David Bussard, David Farrar, Nina Ching Y. Wang, and Paul White for helpful comments during the preparation of this article. In addition, the three anonymous reviewers provided comments that significantly strengthened this article. This work is dedicated to the memory of Fred Power (1938-2007). His keen analytical mind will be greatly missed, but his gentle heart and big smile will be missed even more.

References (131)

  • GreenT. et al.

    Trichloroethylene-induced mouse lung tumors: studies of the mode of action and comparisons between species

    Fundam. Appl. Toxicol.

    (1997)
  • GreenbergM.S. et al.

    Physiologically based pharmacokinetic modeling of inhaled trichloroethylene and its oxidative metabolites in B6C3F1 mice

    Toxicol. Appl. Pharmacol.

    (1999)
  • HissinkE.M. et al.

    The use of in vitro metabolic parameters and physiologically based pharmacokinetic (PBPK) modeling to explore the risk assessment of trichloroethylene

    Environ. Toxicol. Pharmacol.

    (2002)
  • KanekoT. et al.

    Relationship between blood/air partition coefficients of lipophilic organic solvents and blood triglyceride levels

    Toxicology

    (2000)
  • KirmanC.R. et al.

    Assessing the dose-dependency of allometric scaling performance using physiologically based pharmacokinetic modeling

    Regul. Toxicol. Pharmacol.

    (2003)
  • LarsonJ.L. et al.

    Metabolism and lipoperoxidative activity of trichloroacetate and dichloroacetate in rats and mice

    Toxicol. Appl. Pharmacol.

    (1992)
  • LarsonJ.L. et al.

    Species differences in the metabolism of trichloroethylene to the carcinogenic metabolites trichloroacetate and dichloroacetate

    Toxicol. Appl. Pharmacol.

    (1992)
  • LashL.H. et al.

    Susceptibility of primary cultures of proximal tubular and distal tubular cells from rat kidney to chemically induced toxicity

    Toxicology

    (1995)
  • LashL.H. et al.

    Cellular and subcellular heterogeneity of glutathione metabolism and transport in rat kidney cells

    Toxicology

    (1998)
  • LeeK.M. et al.

    Characterization of presystemic elimination of trichloroethylene and its nonlinear kinetics in rats

    Toxicol. Appl. Pharmacol.

    (1996)
  • LeeK.M. et al.

    Contribution of direct solvent injury to the dose-dependent kinetics of trichloroethylene: portal vein administration to rats

    Toxicol. Appl. Pharmacol.

    (2000)
  • LeeK.M. et al.

    Mechanisms of the dose-dependent kinetics of trichloroethylene: oral bolus dosing of rats

    Toxicol. Appl. Pharmacol.

    (2000)
  • LipscombJ.C. et al.

    Cytochrome P450-dependent metabolism of trichloroethylene: interindividual differences in humans

    Toxicol. Appl. Pharmacol.

    (1997)
  • LipscombJ.C. et al.

    In vitro to in vivo extrapolation for trichloroethylene metabolism in humans

    Toxicol. Appl. Pharmacol.

    (1998)
  • MarinoD.J. et al.

    Revised assessment of cancer risk to dichloromethane: part I Bayesian PBPK and dose–response modeling in mice

    Regul. Toxicol. Pharmacol.

    (2006)
  • MerdinkJ.L. et al.

    The extent of dichloroacetate formation from trichloroethylene, chloral hydrate, trichloroacetate, and trichloroethanol in B6C3F1 mice

    Toxicol. Sci.

    (1998)
  • MerdinkJ.L. et al.

    Kinetics of chloral hydrate and its metabolites in male human volunteers

    Toxicology

    (2008)
  • ATSDR, 1997. Toxicological profile for trichloroethylene. Agency for Toxic Substance and Disease Registry....
  • ATSDR, 2003. ToxFAQs for trichloroethylene. Agency for Toxic Substance and Disease Registry....
  • AbbasR.R. et al.

    Pharmacokinetic analysis of chloral hydrate and its metabolism in B6C3F1 mice

    Drug Metab. Dispos.

    (1996)
  • AbbasR. et al.

    Determination of kinetic rate constants for chloral hydrate, trichloroethanol, trichloroacetic acid, and dichloroacetic acid—a physiologically based modeling approach

    Toxicologist

    (1997)
  • AllenB.C. et al.

    Pharmacokinetic modeling of trichloroethylene and trichloroacetic acid in humans

    Risk Anal.

    (1993)
  • AndersM.W. et al.

    Biosynthesis and biotransformation of glutathione S-conjugates to toxic metabolites

    Crit. Rev. Toxicol.

    (1988)
  • BarterZ.E. et al.

    Scaling factors for the extrapolation of in vivo metabolic drug clearance from in vitro data: reaching a consensus on values of human microsomal protein and hepatocellularity per gram of liver

    Curr. Drug Metab.

    (2007)
  • BartonH.A. et al.

    Characterizing uncertainty and variability in physiologically based pharmacokinetic models: state of the science and needs for research and implementation

    Toxicol. Sci.

    (2007)
  • BartonicekV.

    Metabolism and excretion of trichloroethylene after inhalation by human subjects

    Brit. J. Industr. Med.

    (1962)
  • BernauerU. et al.

    Biotransformation of trichloroethene: dose-dependent excretion of 2,2,2-trichloro-metabolites and mercapturic acids in rats and humans after inhalation

    Arch. Toxikol.

    (1996)
  • BernillonP. et al.

    Statistical issues in toxicokinetic modeling: a Bayesian perspective

    Environ. Health Perspect.

    (2000)
  • BirnerG. et al.

    Nephrotoxic and genotoxic N-acetyl-S-dichlorovinyl-l-cysteine is a urinary metabolite after occupational 1,1,2-trichloroethene exposure in humans: implications for the risk of trichloroethene exposure

    Environ. Health Perspect.

    (1993)
  • BloemenL.J. et al.

    Study on the cytochrome P-450-and glutathione-dependent biotransformation of trichloroethylene in humans

    Int. Arch. Occup. Environ. Health

    (2001)
  • BoisF.Y.

    Statistical analysis of Fisher et al. PBPK model of trichloroethylene kinetics

    Environ. Health Perspect.

    (2000)
  • BoisF.Y.

    Statistical analysis of Clewell et al. PBPK model of trichloroethylene kinetics

    Environ. Health Perspect.

    (2000)
  • BoyesW.K. et al.

    Duration adjustment of acute exposure guideline level values for trichloroethylene using a physiologically-based pharmacokinetic model

    Risk Anal.

    (2005)
  • Bronley-DeLanceyA. et al.

    Application of cryopreserved human hepatocytes in trichloroethylene risk assessment: relative disposition of chloral hydrate to trichloroacetate and trichloroethanol

    Environ. Health Perspect.

    (2006)
  • BrownR.P. et al.

    Physiological parameter values for physiologically based pharmacokinetic models

    Toxicol. Ind. Health

    (1997)
  • BrüningT. et al.

    Renal cell cancer risk and occupational exposure to trichloroethylene: results of a consecutive case-control study in Arnsberg, Germany

    Am. J. Ind. Med.

    (2003)
  • BubenJ.A. et al.

    Delineation of the role of metabolism in the hepatotoxicity of trichloroethylene and perchloroethylene: a dose-effect study

    Toxicol. Appl. Pharmacol.

    (1985)
  • BullR.J.

    Mode of action of liver tumor induction by trichloroethylene and its metabolites, trichloroacetate and dichloroacetate

    Environ. Health Perspect.

    (2000)
  • CaiH. et al.

    . Mechanism of aqueous decomposition of trichloroethylene oxide

    J. Am. Chem. Soc.

    (1999)
  • CharbotelB. et al.

    Case-control study on renal cell cancer and occupational exposure to trichloroethylene. Part II: epidemiological aspects

    Ann. Occup. Hyg.

    (2006)
  • Cited by (68)

    • Toxicokinetics of phytonutrients

      2023, Phytonutrients and Neurological Disorders: Therapeutic and Toxicological Aspects
    View all citing articles on Scopus
    1

    Current address: PO Box 16084, Irvine, CA 92623-6084.

    2

    Current address: National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.

    3

    Fax: +1 919 541 4284.

    View full text