1
|
Liu R, Zacharewski TR, Conolly RB, Zhang Q. A Physiologically Based Pharmacokinetic (PBPK) Modeling Framework for Mixtures of Dioxin-like Compounds. TOXICS 2022; 10:toxics10110700. [PMID: 36422908 PMCID: PMC9698634 DOI: 10.3390/toxics10110700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 06/08/2023]
Abstract
Humans are exposed to persistent organic pollutants, such as dioxin-like compounds (DLCs), as mixtures. Understanding and predicting the toxicokinetics and thus internal burden of major constituents of a DLC mixture is important for assessing their contributions to health risks. PBPK models, including dioxin models, traditionally focus on one or a small number of compounds; developing new or extending existing models for mixtures often requires tedious, error-prone coding work. This lack of efficiency to scale up for multi-compound exposures is a major technical barrier toward large-scale mixture PBPK simulations. Congeners in the DLC family, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), share similar albeit quantitatively different toxicokinetic and toxicodynamic properties. Taking advantage of these similarities, here we reported the development of a human PBPK modeling framework for DLC mixtures that can flexibly accommodate an arbitrary number of congeners. Adapted from existing TCDD models, our mixture model contains the blood and three diffusion-limited compartments-liver, fat, and rest of the body. Depending on the number of congeners in a mixture, varying-length vectors of ordinary differential equations (ODEs) are automatically generated to track the tissue concentrations of the congeners. Shared ODEs are used to account for common variables, including the aryl hydrocarbon receptor (AHR) and CYP1A2, to which the congeners compete for binding. Binary and multi-congener mixture simulations showed that the AHR-mediated cross-induction of CYP1A2 accelerates the sequestration and metabolism of DLC congeners, resulting in consistently lower tissue burdens than in single exposure, except for the liver. Using dietary intake data to simulate lifetime exposures to DLC mixtures, the model demonstrated that the relative contributions of individual congeners to blood or tissue toxic equivalency (TEQ) values are markedly different than those to intake TEQ. In summary, we developed a mixture PBPK modeling framework for DLCs that may be utilized upon further improvement as a quantitative tool to estimate tissue dosimetry and health risks of DLC mixtures.
Collapse
Affiliation(s)
- Rongrui Liu
- Lower Merion High School, Ardmore, PA 19003, USA
| | - Tim R. Zacharewski
- Department of Biochemistry and Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
2
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Fürst P, Håkansson H, Halldorsson T, Lundebye AK, Pohjanvirta R, Rylander L, Smith A, van Loveren H, Waalkens-Berendsen I, Zeilmaker M, Binaglia M, Gómez Ruiz JÁ, Horváth Z, Christoph E, Ciccolallo L, Ramos Bordajandi L, Steinkellner H, Hoogenboom LR. Risk for animal and human health related to the presence of dioxins and dioxin-like PCBs in feed and food. EFSA J 2018; 16:e05333. [PMID: 32625737 PMCID: PMC7009407 DOI: 10.2903/j.efsa.2018.5333] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The European Commission asked EFSA for a scientific opinion on the risks for animal and human health related to the presence of dioxins (PCDD/Fs) and DL-PCBs in feed and food. The data from experimental animal and epidemiological studies were reviewed and it was decided to base the human risk assessment on effects observed in humans and to use animal data as supportive evidence. The critical effect was on semen quality, following pre- and postnatal exposure. The critical study showed a NOAEL of 7.0 pg WHO2005-TEQ/g fat in blood sampled at age 9 years based on PCDD/F-TEQs. No association was observed when including DL-PCB-TEQs. Using toxicokinetic modelling and taking into account the exposure from breastfeeding and a twofold higher intake during childhood, it was estimated that daily exposure in adolescents and adults should be below 0.25 pg TEQ/kg bw/day. The CONTAM Panel established a TWI of 2 pg TEQ/kg bw/week. With occurrence and consumption data from European countries, the mean and P95 intake of total TEQ by Adolescents, Adults, Elderly and Very Elderly varied between, respectively, 2.1 to 10.5, and 5.3 to 30.4 pg TEQ/kg bw/week, implying a considerable exceedance of the TWI. Toddlers and Other Children showed a higher exposure than older age groups, but this was accounted for when deriving the TWI. Exposure to PCDD/F-TEQ only was on average 2.4- and 2.7-fold lower for mean and P95 exposure than for total TEQ. PCDD/Fs and DL-PCBs are transferred to milk and eggs, and accumulate in fatty tissues and liver. Transfer rates and bioconcentration factors were identified for various species. The CONTAM Panel was not able to identify reference values in most farm and companion animals with the exception of NOAELs for mink, chicken and some fish species. The estimated exposure from feed for these species does not imply a risk.
Collapse
|
3
|
Filser JG, Baur C, Csan Ädy A, Kessler W, Kreuzer PE. Toxicokinetic Modeling as a Tool for Risk Estimation: 2,3,7,8-Tetrachlorodibenzo-P-Dioxin. Int J Toxicol 2016. [DOI: 10.1080/109158197227053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Concepts of toxicokinetic modeling and the relevance of toxicokinetics for understanding dose-response relationships, species scaling, and risk estimation are broached. A physiological one-compartment model for 2,3,7,8-tetra-chlorodibenzo-p-dioxin (TCDD) is presented in detail. It describes the TCDD burden of the human body, which results from TCDD-contaminated food, in dependence of age. The model was validated using a series of measured values obtained by other authors and this group. They represent lipid-based concentrations of TCDD in liver, blood, adipose tissue, feces, and mother's milk in dependence of age. Special attention was paid to the TCDD burden in infants resulting from feeding with mother's milk or formula. Model simulations demonstrate that TCDD burden can amount to 10 mg/kg of lipids after nursing for 6 months with mother's milk exclusively This is still within the range of the concentrations found in adults. After the nursing period, TCDD burden declines. From the age of 7 years on, there is no longer a difference in the TCDD burden, independently of the food they had received as infants. According to the model, elimination half-life of TCDD from the body is not constant but increases during life starting from a few months in newborns to several years in adults.
Collapse
Affiliation(s)
- J. G. Filser
- GSF—Institut für Toxikologie, Neuherberg, Germany
| | - C. Baur
- Institut für Gerichtsmedizin, Ludwig-Maximilians-Universität München, München, Germany
| | - A. Csan Ädy
- GSF—Institut für Toxikologie, Neuherberg, Germany, and Central Research Institute of Chemistry, Hungarian Academy of Sciences, Budapest, Hungary
| | - W. Kessler
- GSF—Institut für Toxikologie, Neuherberg, Germany
| | | |
Collapse
|
4
|
A Multicompartment Liver-based Pharmacokinetic Model for Benzene and its Metabolites in Mice. Bull Math Biol 2009; 72:507-40. [DOI: 10.1007/s11538-009-9459-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 09/14/2009] [Indexed: 11/25/2022]
|
5
|
Klanjscek T, Nisbet RM, Caswell H, Neubert MG. A model for energetics and bioaccumulation in marine mammals with applications to the right whale. ECOLOGICAL APPLICATIONS : A PUBLICATION OF THE ECOLOGICAL SOCIETY OF AMERICA 2007; 17:2233-50. [PMID: 18213965 DOI: 10.1890/06-0426.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We present a dynamic energy budget (DEB) model for marine mammals, coupled with a pharmacokinetic model of a lipophilic persistent toxicant. Inputs to the model are energy availability and lipid-normalized toxicant concentration in the environment. The model predicts individual growth, reproduction, bioaccumulation, and transfer of energy and toxicant from mothers to their young. We estimated all model parameters for the right whale; with these parameters, reduction in energy availability increases the age at first parturition, increases intervals between reproductive events, reduces the organisms' ability to buffer seasonal fluctuations, and increases its susceptibility to temporal shifts in the seasonal peak of energy availability. Reduction in energy intake increases bioaccumulation and the amount of toxicant transferred from mother to each offspring. With high energy availability, the toxicant load of offspring decreases with birth order. Contrary to expectations, this ordering may be reversed with lower energy availability. Although demonstrated with parameters for the right whale, these relationships between energy intake and energetics and pharmacokinetics of organisms are likely to be much more general. Results specific to right whales include energy assimilation estimates for the North Atlantic and southern right whale, influences of history of energy availability on reproduction, and a relationship between ages at first parturition and calving intervals. Our model provides a platform for further analyses of both individual and population responses of marine mammals to pollution, and to changes in energy availability, including those likely to arise through climate change.
Collapse
Affiliation(s)
- Tin Klanjscek
- Biology Department, MS#34, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.
| | | | | | | |
Collapse
|
6
|
Yokley K, Tran HT, Pekari K, Rappaport S, Riihimaki V, Rothman N, Waidyanatha S, Schlosser PM. Physiologically-based pharmacokinetic modeling of benzene in humans: a Bayesian approach. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2006; 26:925-43. [PMID: 16948686 DOI: 10.1111/j.1539-6924.2006.00789.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Benzene is myelotoxic and leukemogenic in humans exposed at high doses (>1 ppm, more definitely above 10 ppm) for extended periods. However, leukemia risks at lower exposures are uncertain. Benzene occurs widely in the work environment and also indoor air, but mostly below 1 ppm, so assessing the leukemia risks at these low concentrations is important. Here, we describe a human physiologically-based pharmacokinetic (PBPK) model that quantifies tissue doses of benzene and its key metabolites, benzene oxide, phenol, and hydroquinone after inhalation and oral exposures. The model was integrated into a statistical framework that acknowledges sources of variation due to inherent intra- and interindividual variation, measurement error, and other data collection issues. A primary contribution of this work is the estimation of population distributions of key PBPK model parameters. We hypothesized that observed interindividual variability in the dosimetry of benzene and its metabolites resulted primarily from known or estimated variability in key metabolic parameters and that a statistical PBPK model that explicitly included variability in only those metabolic parameters would sufficiently describe the observed variability. We then identified parameter distributions for the PBPK model to characterize observed variability through the use of Markov chain Monte Carlo analysis applied to two data sets. The identified parameter distributions described most of the observed variability, but variability in physiological parameters such as organ weights may also be helpful to faithfully predict the observed human-population variability in benzene dosimetry.
Collapse
Affiliation(s)
- Karen Yokley
- Department of Mathematics and Center for Research in Scientific Computation, North Carolina State University, Raleigh, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Abstract
In recent years physiologically based pharmacokinetic (PBPK) modeling has found frequent application in risk assessments where PBPK models serve as important adjuncts to studies on modes of action of xenobiotics. In this regard, studies on mode of action provide insight into both the sites/mechanisms of action and the form of the xenobiotic associated with toxic responses. Validated PBPK models permit calculation of tissue doses of xenobiotics and metabolites for a variety of conditions, i.e. at low-doses, in different animal species, and in different members of a human population. In this manner, these PBPK models support the low-dose and interspecies extrapolations that are important components of current risk assessment methodologies. PBPK models are sometimes referred to as physiological toxicokinetic (PT) models to emphasize their application with compounds causing toxic responses. Pharmacokinetic (PK) modeling in general has a rich history. Data-based PK compartmental models were developed in the 1930's when only primitive tools were available for solving sets of differential equations. These models were expanded in the 1960's and 1970's to accommodate new observations on dose-dependent elimination and flow-limited metabolism. The application of clearance concepts brought many new insights about the disposition of drugs in the body. In the 1970's PBPK/PT models were developed to evaluate metabolism of volatile compounds of occupational importance, and, for the first time, dose-dependent processes in toxicology were included in PBPK models in order to assess the conditions under which saturation of metabolic and elimination processes lead to non-linear dose response relationships. In the 1980's insights from chemical engineers and occupational toxicology were combined to develop PBPK/PT models to support risk assessment with methylene chloride and other solvents. The 1990's witnessed explosive growth in risk assessment applications of PBPK/PT models and in applying sensitivity and variability methods to evaluate model performance. Some of the compounds examined in detail include butadiene, styrene, glycol ethers, dioxins and organic esters/aids. This paper outlines the history of PBPK/PT modeling, emphasizes more recent applications of PBPK/TK models in health risk assessment, and discusses the risk assessment perspective provided by modern uses of these modeling approaches.
Collapse
Affiliation(s)
- Melvin E Andersen
- Department of Environmental Health, International Center for Risk Assessment, Environmental Health Building, Colorado State University, Ft. Collins, CO 80523-1860, USA.
| |
Collapse
|
8
|
Miniero R, De Felip E, Ferri F, di Domenico A. An overview of TCDD half-life in mammals and its correlation to body weight. CHEMOSPHERE 2001; 43:839-844. [PMID: 11372874 DOI: 10.1016/s0045-6535(00)00442-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Major determinants of TCDD half-life in organisms are lipophilicity, metabolism, and hepatic binding sites. In addition, half-life seems to be empirically correlated to organism body weight. In this paper, this correlation is evaluated by a regression analysis of half-life measures and body weight data selected from the literature. Single exposure studies on laboratory mammals and human half-life data were specifically taken in consideration. The analysis outcome appears to be highly significant probably owing to the stability and generally slow metabolism of the substance in the organisms considered. The effect on half-life of factors other than body weight does not seem to influence significantly data dispersion around the regression line. The potential effects of a dose-dependent excretion cannot be excluded as toxicokinetic studies have been usually carried out at high exposure doses.
Collapse
Affiliation(s)
- R Miniero
- Laboratory of Comparative Toxicology and Ecotoxicology, Instituto Superiore di Sanità, Rome, Italy
| | | | | | | |
Collapse
|
9
|
Poellinger L. Mechanistic aspects--the dioxin (aryl hydrocarbon) receptor. FOOD ADDITIVES AND CONTAMINANTS 2000; 17:261-6. [PMID: 10912240 DOI: 10.1080/026520300283333] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The Ah receptor mediates the toxicological responses of 2,3,7,8-TCDD and related compounds. Receptor-deficient animals were shown to be resistant to the toxic effects of dioxin, although there is also evidence for the existence of a receptor-independent pathway for dioxin-induced toxicity. In the cytosol the receptor is present in a non-activated ligand binding conformation. Association with Arnt in the nucleus turns the receptor complex into a ligand activated form. The physiological role of the receptor is not yet understood; however, the conservation of the receptor in a wide range of animal species (including humans) suggests a fundamental role in cellular physiology.
Collapse
Affiliation(s)
- L Poellinger
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
Santostefano MJ, Wang X, Richardson VM, Ross DG, DeVito MJ, Birnbaum LS. A pharmacodynamic analysis of TCDD-induced cytochrome P450 gene expression in multiple tissues: dose- and time-dependent effects. Toxicol Appl Pharmacol 1998; 151:294-310. [PMID: 9707506 DOI: 10.1006/taap.1998.8466] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ability of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) to alter gene expression and the demonstration that the induction of CYP1A2 is responsible for hepatic TCDD sequestration suggest that both pharmacokinetic and pharmacodynamic events must be incorporated for a quantitative description of TCDD disposition. In this paper, a biologically based pharmacodynamic (BBPD) model for TCDD-induced biochemical responses in multiple tissues was developed. The parameters responsible for tissue response were estimated simultaneously with a refined physiologically based pharmacokinetic (PBPK) model developed by Wang et al. (1997a), by using the time-dependent effects of TCDD on induced CYP1A1/CYP1A2 gene expression in multiple target tissues (liver, lungs, kidneys, and skin) of female Sprague-Dawley rats treated with 10 microgram TCDD/kg for 30 min, 1, 3, 8, or 24 h, or 7, 14, or 35 days. This refined BBPD model developed based on the time-course of TCDD-induced CYP1A1/CYP1A2 protein expression, and associated enzymatic activities well described the dose-dependent effects of TCDD on cytochrome P450 protein expression and associated enzyme activities in the multiple tissues of female Sprague-Dawley rats at 3 days following a single exposure to TCDD (0.01-30.0 micromgram TCDD/kg). This is the first BBPD model to quantitatively describe the time- and dose-dependent effects of TCDD on induced CYP1A1/CYP1A2 protein expression and associated enzyme activities in multiple target tissues for TCDD-induced biochemical responses.
Collapse
Affiliation(s)
- M J Santostefano
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, North Carolina 27599-7270, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Lilly PD, Andersen ME, Ross TM, Pegram RA. Physiologically based estimation of in vivo rates of bromodichloromethane metabolism. Toxicology 1997; 124:141-52. [PMID: 9458004 DOI: 10.1016/s0300-483x(97)00146-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bromodichloromethane (BDCM) is a rodent carcinogen formed by chlorination of drinking water containing bromide and organic precursors. BDCM is a member of the class of disinfection by-products known as trihalomethanes (THMs), compounds that have been shown to be carcinogenic in rodents. A physiologically-based pharmacokinetic (PBPK) model has been developed and applied to provide estimates of the rates of metabolism of BDCM in vivo in rats. The model consists of five compartments (liver, kidney, fat and slowly and rapidly perfused tissues). Tissue partition coefficients were determined using a modified vial equilibration technique and rates of metabolism were estimated by fitting data obtained from stable metabolite (bromide ion, (Br-)) analysis following 4 h constant concentration BDCM inhalation exposure (50-3200 ppm) and closed chamber gas uptake experiments. Metabolism was described using a single saturable pathway representing a high capacity, high affinity process (Vmaxc = 12.8 mg/h/kg; Km = 0.5 mg/l). Rate constants obtained from Br- data adequately described data from gas uptake experiments and literature data on exhalation of 14CO and 14CO2 produced following oral gavage with 14C-BDCM. Pretreatment with trans-dichloroethylene (t-DCE), an inhibitor of CYP2E1, increased the apparent Km from 0.5 to 225 mg/l indicating that CYP2E1 is the major P450 isoform involved in the bioactivation of BDCM to reactive intermediates.
Collapse
Affiliation(s)
- P D Lilly
- Experimental Toxicology Division, MD-74, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | | | | | | |
Collapse
|
12
|
Wang X, Santostefano MJ, Evans MV, Richardson VM, Diliberto JJ, Birnbaum LS. Determination of parameters responsible for pharmacokinetic behavior of TCDD in female Sprague-Dawley rats. Toxicol Appl Pharmacol 1997; 147:151-68. [PMID: 9356318 DOI: 10.1006/taap.1997.8242] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is the most toxic member of a class of planar and halogenated chemicals. Improvements in exposure assessment of TCDD require scientific information on the distribution of TCDD in target tissues and cellular responses induced by TCDD. Since 1980, several physiologically based pharmacokinetic (PBPK) models for TCDD and related compounds have been reported. Some of these models incorporated the induction of a hepatic binding protein in response to interactions of TCDD, the Ah receptor, and DNA binding sites and described the TCDD disposition in a biological system for certain data sets. Due to the limitations of the available experimental data, different values for the same physical parameters of these models were obtained from the different studies. The inconsistencies of the parameter values limit the application of PBPK models to risk assessment. Therefore, further refinement of previous models is necessary. This paper develops an improved PBPK model to describe TCDD disposition in eight target tissues. The interaction of TCDD with the Ah receptor and with hepatic inducible CYP1A2 were also incorporated into the model. This model accurately described the time course distribution of TCDD following a single oral dose of 10 microg/kg, as well as the TCDD concentration on Day 3 after six different doses, 0.01, 0.1, 0.3, 1, 10, and 30 microg TCDD/kg, in target tissues. This study extends previous TCDD models by illustrating the validity and the limitation of the model and providing further confirmation of the potential PBPK model for us in optimal experimental design and extrapolation across doses and routes of exposure. In addition, this study demonstrated some critical issues in PBPK modeling.
Collapse
Affiliation(s)
- X Wang
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, North Carolina 27599-7270, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Lawrence GS, Gobas FA. A pharmacokinetic analysis of interspecies extrapolation in dioxin risk assessment. CHEMOSPHERE 1997; 35:427-452. [PMID: 9241819 DOI: 10.1016/s0045-6535(97)00108-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
This study entails a pharmacokinetic analysis of the relationship between the external dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin, TCDD) and resulting concentrations of TCDD in internal tissues and organs of humans and rodent species. The methodology is based on the development and testing of physiologically based pharmacokinetic models for several rodent species and humans. The results indicate that the relationship between the external dose of TCDD and resulting TCDD concentrations in liver and adipose tissue of humans and various species of rats and mice can vary by as much as 725 fold, illustrating that humans and experimental animals differ considerably in their ability to convert external dosages of dioxin to tissue concentrations. Interspecies scaling factors are reported to express the differences in tissue concentrations of dioxin between mice, rats and humans in response to an equivalent external dose. The significance of these findings for conducting human cancer and ecological risk assessments is discussed. It is recommended that pharmacokinetic differences be considered explicity in risk estimation, while separately recognizing interspecies differences in pharmacodynamics (sensitivity).
Collapse
Affiliation(s)
- G S Lawrence
- School of Resource and Environmental Management, Simon Fraser University Burnaby, British Columbia, Canada
| | | |
Collapse
|
14
|
Andersen ME, Birnbaum LS, Barton HA, Eklund CR. Regional hepatic CYP1A1 and CYP1A2 induction with 2,3,7,8-tetrachlorodibenzo-p-dioxin evaluated with a multicompartment geometric model of hepatic zonation. Toxicol Appl Pharmacol 1997; 144:145-55. [PMID: 9169078 DOI: 10.1006/taap.1996.8067] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A physiologically based pharmacokinetic (PBPK) model for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was combined with a five-compartment geometric model of hepatic zonation to predict both total and regional induction of CYP450 proteins within the liver. Three literature studies on TCDD pharmacokinetics and protein induction in female rats were analyzed. In simulating low-dose behavior for mRNA in whole liver and, particularly, in representing immunohistochemical observations, the five-compartment model was more successful than conventional homogeneous one-compartment liver models. The five-compartment liver model was used with the affinity of TCDD for the Ah receptor (AhR) held constant across all the liver (Kb = 0.2 nM). The presumed affinities of the AhR-TCDD complex for TCDD responsive elements in the CYP1A1 (Kd1) and CYP1A2 (Kd2) genes varied between adjacent compartments by a factor of 3. This parameterization leads to predicted 81-fold differences in affinities between the centrilobular and the periportal regions. The affinities used for AhR-TCDD complex binding to TCDD response elements for CYP1A2 in compartment 3 (the midzonal area) ranged from 0.08 to 1.0 nM in the three studies modeled. For CYP1A1 the corresponding dissociation constant in compartment 3 varied from 0.6 to 2.0 nM. In each compartment, the Hill coefficient for induction had to be 4 or greater to match the immunohistochemical results. This multi-compartment liver model is consistent with data on protein and mRNA induction throughout the liver and on the regional distribution of these proteins. No previous model has incorporated regional variations in induction. The PBPK analysis based on the multicompartment liver model suggests that the low-dose behavior for hepatic CYP1A1/CYP1A2 induction by TCDD is highly non-linear.
Collapse
Affiliation(s)
- M E Andersen
- ICF Kaiser Engineers, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | |
Collapse
|
15
|
Hays SM, Aylward LL, Karch NJ, Paustenbach DJ. The relative susceptibility of animals and humans to the carcinogenic hazard posed by exposure to 2,3,7,8-TCDD: an analysis using standard and internal measures of dose. CHEMOSPHERE 1997; 34:1507-1522. [PMID: 9134683 DOI: 10.1016/s0045-6535(97)00447-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
An analysis of the carcinogenic dose-response for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in humans and animals was performed based on measured tissue and serum concentrations and using alternatives to administered dose as the dosimetric. The TCDD-related carcinogenic response in rats (female rat liver tumors from Kociba et al., using revised pathology from Goodman and Sauer, was compared to that in humans (lung cancer rates in Fingerhut et al.,). Three dosimetries were used: serum lipid TCDD area-under-the-curve (AUC), peak serum lipid concentration (Cpeak) and average serum lipid concentration (Cavg). Rat serum concentration-time profiles were estimated based on measured adipose lipid TCDD concentrations at the end of the Kociba et al. bioassay, assuming first-order elimination and a half-life of 25 days. Human concentration-time profiles were estimated based on measured serum lipid TCDD concentrations and known dates of first and last exposure, with an assumed 7.5 year half-life and first-order elimination. Comparison of rat and human responses indicated that, using all three of these dosimetries, humans are much less sensitive than rats to the carcinogenic effects of TCDD. Regardless of the dosimetric chosen, the cancer mortality in humans in the NIOSH cohort, if due to TCDD, is relatively insensitive to dose as defined in Fingerhut et al., [3]. Our analysis indicates that human exposure to background levels of TCDD (about 5 ppt serum lipid concentration) is not likely to produce an incremental cancer risk.
Collapse
Affiliation(s)
- S M Hays
- ChemRisk, A Division of McLaren/Hart, Cleveland, OH 44122, USA
| | | | | | | |
Collapse
|
16
|
Goddard MJ, Krewski D. The future of mechanistic research in risk assessment: where are we going and can we get there from here? Toxicology 1995; 102:53-70. [PMID: 7482562 DOI: 10.1016/0300-483x(95)03036-f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Quantitative estimates of human health risk are often based on mathematical models fit to experimental or epidemiological data. Recent years have witnessed a trend towards the use of mechanistic models in risk assessment applications. Such models afford a more biologically based interpretation of the data and a firmer scientific basis for extrapolation beyond the conditions under which the original data were obtained. In this article, we review some recent advances in the development of biologically based models for mutagenesis, carcinogenesis and developmental toxicity. Pharmacokinetic and receptor-binding models and their roles in mechanistic risk assessment are also discussed. The future of mechanistic research in risk assessment is contemplated, including the need for more elaborate experiments to obtain the data necessary for mechanistic modeling.
Collapse
Affiliation(s)
- M J Goddard
- Environmental Health Centre, Tunney's Pasture, Ottawa, Ontario, Canada
| | | |
Collapse
|
17
|
Abstract
Significant advances have been made in the development of physiologically-based models of dioxin pharmacokinetics (PBPK) in the last 5-6 years. These models incorporate explicit descriptions of biological factors which determine tissue dosimetry of dioxin and include some description of dioxin-mediated pharmacodynamic events. Biological determinants of dioxin disposition include fat solubility, specific and inducible binding in the liver, diffusion-limited tissue distribution and metabolic elimination. PBPK models have been successfully used to predict the dose and time-dependent chemical disposition and protein induction properties of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) over a wide variety of experimental data sets with rodents. The models have also been extended to describe the disposition of a brominated dioxin, 2,3,7,8-tetrabromodibenzo-p-dioxin. As these quantitative descriptions of disposition are more fully refined, particularly with regard to pharmacodynamic descriptions of dioxin-mediated alterations in gene expression, more accurate predictions of tissue dosimetry and tissue responses will be performed across dose, species and related polyhalogenated aromatic hydrocarbons. Accurate, mechanistic dosimetry models will facilitate biologically-based approaches to the human risk assessment of these important and ubiquitous environmental contaminants.
Collapse
Affiliation(s)
- L A Buckley
- Eli Lilly and Company, Toxicology Research Laboratories, Greenfield, IN 46140, USA
| |
Collapse
|
18
|
Leung HW, Paustenbach DJ. Physiologically based pharmacokinetic and pharmacodynamic modeling in health risk assessment and characterization of hazardous substances. Toxicol Lett 1995; 79:55-65. [PMID: 7570674 DOI: 10.1016/0378-4274(95)03357-q] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Recent advances in physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) modeling have introduced novel approaches for evaluating toxicological problems. Because PBPK models are amenable to extrapolation of tissue dosimetry, they are increasingly being applied to chemical risk assessment. A comprehensive listing of PBPK/PD models for environmental chemicals developed to date is referenced. Salient applications of PBPK/PD modeling to health risk assessments and characterization of hazardous substances are illustrated with examples.
Collapse
Affiliation(s)
- H W Leung
- Union Carbide Corporation, Danbury, CT 06817, USA
| | | |
Collapse
|
19
|
Van den Berg M, De Jongh J, Poiger H, Olson JR. The toxicokinetics and metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs) and their relevance for toxicity. Crit Rev Toxicol 1994; 24:1-74. [PMID: 8172651 DOI: 10.3109/10408449409017919] [Citation(s) in RCA: 285] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
This article reviews the present state of the art regarding the toxicokinetics and metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs). The absorption, body distribution, and metabolism can vary greatly between species and also may depend on the congener and dose. In biota, the 2,3,7,8-substituted PCDDs and PCDFs are almost exclusively retained in all tissue types, preferably liver and fat. This selective tissue retention and bioaccumulation are caused by a reduced rate of biotransformation and subsequent elimination of congeners with chlorine substitution at the 2,3,7, and 8 positions. 2,3,7,8-Substituted PCDDs and PCDFs also have the greatest toxic and biological activity and affinity for the cytosolic arylhydrocarbon (Ah)-receptor protein. The parent compound is the causal agent for Ah-receptor-mediated toxic and biological effects, with metabolism and subsequent elimination of 2,3,7,8- substituted congeners representing a detoxification process. Congener-specific affinity of PCDDs and PCDFs for the Ah-receptor, the genetic events following receptor binding, and toxicokinetics are factors that contribute to the relative in vivo potency of an individual PCDD or PCDF in a given species. Limited human data indicate that marked species differences exist in the toxicokinetics of these compounds. Thus, human risk assessment for PCDDs and PCDFs needs to consider species-, congener-, and dose-specific toxicokinetic data. In addition, exposure to complex mixtures, including PCBs, has the potential to alter the toxicokinetics of individual compounds. These alterations in toxicokinetics may be involved in some of the nonadditive toxic or biological effects that are observed after exposure to mixtures of PCDDs or PCDFs with PCBs.
Collapse
Affiliation(s)
- M Van den Berg
- Research Institute of Toxicology, Utrecht University, The Netherlands
| | | | | | | |
Collapse
|
20
|
Lin WQ, White KL. Modulation of liver intracellular C3 in mice by 2,3,7,8-tetrachlorodibenzo-p-dioxin. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH 1993; 39:107-19. [PMID: 8492324 DOI: 10.1080/15287399309531739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Earlier studies from this laboratory have shown that the complement system, especially the component C3, in female B6C3F1 mice is suppressed following TCDD exposure in vivo. However, the direct exposure of TCDD in vitro does not affect the C3-producing capacity of two types of hepatoma cells, as well as mouse primary hepatocytes. To investigate the effect of TCDD on C3 production by the liver following in vivo exposure, liver intracellular C3 levels and pro-C3, the precursor of the secreted C3, were examined in the present study. The results demonstrated that there was a dose-dependent increase of liver intracellular C3 levels (from 138% to 175% of control) immediately following TCDD (from 10 to 40 micrograms/kg) exposure. This increase was rapid (4 h after exposure), but transient (less than 2 h), and was not accompanied by an alteration of serum C3 levels. Studies using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis showed that the increase in liver intracellular C3 levels resulted from, at least partially, an increase in intracellular pro-C3. Serum C3 levels did not decrease until d 3 after exposure, when both liver intracellular C3 levels and pro-C3 in TCDD-treated mice were not different from those of the control mice. These results indicated that the modulation of liver intracellular C3 by TCDD did not correlate with the suppression of serum C3 levels following exposure.
Collapse
Affiliation(s)
- W Q Lin
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond 23298-0613
| | | |
Collapse
|
21
|
Abstract
There has been considerable progress in recent years in developing physiological models for the pharmacokinetics of toxic chemicals and in the application of these models in cancer risk assessment. Physiological pharmacokinetic models consist of a number of individual compartments, based on the anatomy and physiology of the mammalian organism of interest, and include specific parameters for metabolism, tissue binding, and tissue reactivity. Because of the correspondence between these compartments and specific tissues or groups of tissues, these models are particularly useful for predicting the doses of biologically active forms of toxic chemicals at target tissues under a wide variety of exposure conditions and in different animal species, including humans. Due to their explicit characterization of the biological processes governing pharmacokinetic behaviour, these models permit more accurate predictions of the dose of active metabolites reaching target tissues in exposed humans and hence of potential cancer risk. In addition, physiological models also permit a more direct evaluation of the impact of parameter uncertainty and inter-individual variability in cancer risk assessment. In this article, we review recent developments in physiologic pharmacokinetic modeling for selected chemicals and the application of these models in carcinogenic risk assessment. We examine the use of these models in integrating diverse information on pharmacokinetics and pharmacodynamics and discuss challenges in extending these pharmacokinetic models to reflect more accurately the biological events involved in the induction of cancer by different chemicals.
Collapse
Affiliation(s)
- M E Andersen
- Duke University Medical Center, Durham, NC 27710
| | | | | |
Collapse
|
22
|
Andersen ME, Mills JJ, Gargas ML, Kedderis L, Birnbaum LS, Neubert D, Greenlee WF. Modeling receptor-mediated processes with dioxin: implications for pharmacokinetics and risk assessment. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 1993; 13:25-36. [PMID: 8383868 DOI: 10.1111/j.1539-6924.1993.tb00726.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin; TCDD), a widespread polychlorinated aromatic hydrocarbon, caused tumors in the liver and other sites when administered chronically to rats at doses as low as 0.01 microgram/kg/day. It functions in combination with a cellular protein, the Ah receptor, to alter gene regulation, and this resulting modulation of gene expression is believed to be obligatory for both dioxin toxicity and carcinogenicity. The U.S. EPA is reevaluating its dioxin risk assessment and, as part of this process, will be developing risk assessment approaches for chemicals, such as dioxin, whose toxicity is receptor-mediated. This paper describes a receptor-mediated physiologically based pharmacokinetic (PB-PK) model for the tissue distribution and enzyme-inducing properties of dioxin and discusses the potential role of these models in a biologically motivated risk assessment. In this model, ternary interactions among the Ah receptor, dioxin, and DNA binding sites lead to enhanced production of specific hepatic proteins. The model was used to examine the tissue disposition of dioxin and the induction of both a dioxin-binding protein (presumably, cytochrome P4501A2), and cytochrome P4501A1. Tumor promotion correlated more closely with predicted induction of P4501A1 than with induction of hepatic binding proteins. Although increased induction of these proteins is not expected to be causally related to tumor formation, these physiological dosimetry and gene-induction response models will be important for biologically motivated dioxin risk assessments in determining both target tissue dose of dioxin and gene products and in examining the relationship between these gene products and the cellular events more directly involved in tumor promotion.
Collapse
Affiliation(s)
- M E Andersen
- Chemical Industry Institute of Toxicology, Research Triangle Park, North Carolina 27709
| | | | | | | | | | | | | |
Collapse
|
23
|
Brown MM, McCready TL, Bunce NJ. Factors affecting the toxicity of dioxin-like toxicants: a molecular approach to risk assessment of dioxins. Toxicol Lett 1992; 61:141-7. [PMID: 1322574 DOI: 10.1016/0378-4274(92)90140-f] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The numerous toxic responses of dioxin-like compounds are mediated by the intracellular Ah (aryl hydrocarbon) receptor. It has been suggested that the regulation of dioxins and similar substances could be placed on a molecular foundation by considering the proportion of Ah-receptor sites occupied by toxicant molecules. The present work has shown that the following formation not yet available would be needed in order to develop this approach: correlation between dioxin exposure and human tissue levels; accurate determination of the association constants for human Ah-receptor with toxicant, and for human receptor-ligand complex with DNA; and knowledge of the intracellular concentrations of both receptor binding sites and DNA binding sites. Furthermore, since not all dioxin-like substances behave identically, this information would need to be gathered for a wide variety of substances.
Collapse
Affiliation(s)
- M M Brown
- Department of Chemistry and Biochemistry, University of Guelph, Ontario, Canada
| | | | | |
Collapse
|
24
|
Keenan RE, Paustenbach DJ, Wenning RJ, Parsons AH. Pathology reevaluation of the Kociba et al. (1978) bioassay of 2,3,7,8-TCDD: implications for risk assessment. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH 1991; 34:279-96. [PMID: 1942119 DOI: 10.1080/15287399109531568] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The chronic bioassay of 2,3,7,8-tetrachlorodibenzo-p-dioxin (2,3,7,8-TCDD) reported in 1978 by Kociba et al. has been considered to be the primary evidence supporting its carcinogenicity, and is the basis for most dioxin regulations in North America and Western Europe. Because the histopathological criteria for proliferative lesions in the rat liver have changed significantly since 1978, a reevaluation of the liver slides was conducted recently by an independent panel of pathologists. Using current National Toxicology Program criteria, their study showed, in contrast to the original findings, that about two-thirds fewer tumors were present in the livers of female Sprague-Dawley rats. The no-observed-adverse-effect level (NOAEL) for hepatocellular carcinomas was 0.01 micrograms/kg/d rather than 0.001 micrograms/kg/d, which had been reported in 1978. In light of these significant findings, a quantitative dose-response assessment of 2,3,7,8-TCDD was undertaken to predict the potential carcinogenic risks to humans. Risk-specific doses (RsDs) and cancer potency factors (CPFs) were calculated by applying the linearized multistage (LMS) model to the combined incidences of hepatocellular carcinomas and adenomas, classified in accordance with the 1990 histopathological criteria. Based on the weight of evidence regarding the mechanism of action of 2,3,7,8-TCDD, body weight rather than surface area was selected as the appropriate means for scaling rodent data to predict the human response. Using the survival-adjusted data, the RsD for a 1 in 1,000,000 (10(-6)) plausible upper bound (95%) lifetime incremental cancer risk was 370 fg/kg/d based only on the incidence of hepatocellular carcinomas, and 100 fg/kg/d when hepatocellular carcinomas and adenomas were combined. The corresponding upper-bound (95%) CPFs were 2700 and 9700 (mg/kg/d)-1, respectively. These results indicate that the carcinogenic risk to humans from exposure to 2,3,7,8-TCDD is at least 16-fold lower than previous estimates derived from the Kociba et al. (1978) bioassay.
Collapse
Affiliation(s)
- R E Keenan
- ChemRisk, Division of McLaren/Hart, Portland, ME 04102
| | | | | | | |
Collapse
|
25
|
Affiliation(s)
- J P Landers
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | | |
Collapse
|
26
|
Kedderis LB, Diliberto JJ, Birnbaum LS. Disposition and excretion of intravenous 2,3,7,8-tetrabromodibenzo-p-dioxin (TBDD) in rats. Toxicol Appl Pharmacol 1991; 108:397-406. [PMID: 2020967 DOI: 10.1016/0041-008x(91)90086-t] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Polybrominated dibenzodioxins and dibenzofurans are of toxicologic interest due to potential occupational and environmental exposure and because of their structural similarity to the highly toxic chlorinated analogues. The excretion and terminal tissue distribution of [3H]TBDD was studied in male F344 rats for 56 days following single iv doses of .001 or 0.1 mumol/kg. The major tissue depots of radioactivity were liver, adipose tissue, and skin, and tissue distribution was dose-dependent. At 56 days, liver concentrations in the high dose group were disproportionately increased compared to those of the low dose group. Liver:adipose tissue concentration ratios were 0.2 and 2.6 at the low and high doses, respectively. Elimination of radioactivity in the feces, the major route of excretion, and urine was also nonlinear with respect to dose. By Day 56, feces accounted for approximately 50% of the administered dose at the low dose versus 70% at the high dose. Based on fecal excretion, the apparent terminal whole body half-life was estimated to be 18 days for both dose groups. The time-dependent pattern of tissue disposition was characterized at the low dose over a 56-day period. Blood levels of radioactivity declined rapidly with 2% remaining in the blood by 24 hr. Radioactivity levels in the liver peaked by 7 hr and then gradually declined concomitant with a slow accumulation in adipose tissue. The terminal excretion half-life of radioactivity in adipose tissue was estimated to be 60 days. Liver:adipose tissue concentration ratios declined with time. Thus, the overall disposition of TBDD appears similar to that observed for the chlorinated analogue, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The results of these studies are consistent with the hypothesis that TBDD, like TCDD, induces a binding species in the liver which accounts for higher liver:adipose tissue concentration ratios at the high dose. The dose-dependent tissue disposition and excretion kinetics of these compounds suggest important considerations for extrapolations from high to low doses.
Collapse
Affiliation(s)
- L B Kedderis
- Experimental Toxicology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | | | | |
Collapse
|
27
|
Leung HW. Development and utilization of physiologically based pharmacokinetic models for toxicological applications. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH 1991; 32:247-67. [PMID: 2002511 DOI: 10.1080/15287399109531480] [Citation(s) in RCA: 84] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recent advances in physiologically based pharmacokinetic (PB-PK) modeling have introduced novel approaches for evaluating toxicological problems. Because PB-PK models are amenable to extrapolation of tissue dosimetry, they are increasingly being applied to chemical risk assessment. This paper reviews the development of PB-PK modeling for toxicological applications. It briefly compares and contrasts the fundamental differences between conventional compartmental analysis and PB-PK modeling. The theory and principles, data requirements and the methodologies to obtain them, and the steps to construct PB-PK models are described. A comprehensive listing of PB-PK models for environmental chemicals developed to date is referenced. Salient applications of PB-PK modeling to toxicological problems are illustrated with examples. Finally, the uncertainties and limitations in PB-PK modeling are also discussed.
Collapse
Affiliation(s)
- H W Leung
- Union Carbide Chemicals and Plastics Company, Inc., Health, Safety, & Environmental Affairs, Danbury, Connecticut
| |
Collapse
|
28
|
Leung HW, Paustenbach DJ, Murray FJ, Andersen ME. A physiological pharmacokinetic description of the tissue distribution and enzyme-inducing properties of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat. Toxicol Appl Pharmacol 1990; 103:399-410. [PMID: 2160135 DOI: 10.1016/0041-008x(90)90313-j] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A five-compartment physiologically based pharmacokinetic (PB-PK) model was developed to describe the tissue disposition of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the Sprague-Dawley rat. This description included blood, liver, fat, muscle/skin, and visceral tissue groups. On the basis of other literature, the liver compartment was modeled to include two TCDD-binding sites, corresponding to a cytosolic receptor and a microsomal binding protein. A pharmacodynamic description was developed in which microsomal enzyme induction, both of arylhydrocarbon hydroxylase activity and of the amount of the microsomal TCDD-binding protein, was linked to fractional occupancy of the cytosolic receptor. This description was then used to analyze previously published data on TCDD disposition. The dissociation constant of the cytosolic Ah receptor (KB1) in vivo was estimated to be 15 pM by fitting enzyme induction data from McConnell et al. (1984). The ratio of liver to fat concentration of TCDD (about 4:1) was found to be primarily determined by the dissociation constant of the microsomal binding protein (7 nM) and the basal and induced concentration of this protein in the liver (25 and 200 nmol/liver, respectively). With these parameter values, the tissue distribution of TCDD in fat and liver, the two primary sites of accumulation, was accurately described following either single or repeated dosing with TCDD in the rat. The pharmacokinetic behavior described by the model was extremely sensitive to binding affinities, and only moderately sensitive to binding capacities in the dose range studied. Induction of microsomal TCDD-binding proteins was necessary in order to account for the differences in disposition at low (0.01 microgram/kg) and high (1.0 microgram/kg) daily doses of TCDD. Since the tumorigenicity of TCDD in rats is believed to be correlated with the biological responses of the Ah-TCDD complex, the present physiological pharmacokinetic description, which contains information on receptor occupancy at various dose levels, provides a plausible mechanistic connection for devising pharmacodynamic models which predict the cancer risk of TCDD in the rat.
Collapse
Affiliation(s)
- H W Leung
- Syntex Corporation, Palo Alto, California 94304
| | | | | | | |
Collapse
|