Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Front Pharmacol ; 14: 1223808, 2023.
Article in English | MEDLINE | ID: mdl-37663267

ABSTRACT

Introduction: ß-chloroprene (2-chloro-1,3-butadiene; CP) causes lung tumors after inhalation exposures in rats and mice. Mice develop these tumors at lower exposures than rats. In rats CP exposures cause depletion of lung glutathione (GSH). Methods: PBPK models developed to relate the appearance of mouse lung tumors with rates of CP metabolism to reactive metabolites or total amounts metabolized during exposures have been expanded to include production of reactive metabolites from CP. The extended PBPK model describes both the unstable oxirane metabolite, 2-CEO, and metabolism of the more stable oxirane, 1-CEO, to reactive metabolites via microsomal oxidation to a diepoxide, and linked production of these metabolites to a PK model predicting GSH depletion with increasing CP exposure. Key information required to develop the model were available from literature studies identifying: 1) microsomal metabolites of CP, and 2) in vitro rates of clearance of CP and 1-CEO from active microsomal preparations from mice, rats, hamsters and humans. Results: Model simulation of concentration dependence of disproportionate increases in reactive metabolite concentrations as exposures increases and decreases in tissue GSH are consistent with the dose-dependence of tumor formation. At the middle bioassay concentrations with a lung tumor incidence, the predicted tissue GSH is less than 50% background. These simulations of reduction in GSH are also consistent with the gene expression results showing the most sensitive pathways are Nrf2-regulation of oxidative stress and GSH metabolism. Discussion: The PBPK model is used to correlate predicted tissue exposure to reactive metabolites with toxicity and carcinogenicity of CP.

3.
Toxicol Appl Pharmacol ; 380: 114695, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31394159

ABSTRACT

A previously published human PBPK model for manganese (Mn) in infants and children has been updated with Mn in drinking water as an additional exposure source. Built upon the ability to capture differences in Mn source-specific regulation of intestinal uptake in nursing infants who are breast-fed and formula-fed, the updated model now describes the bioavailability of Mn from drinking water in children of ages 0-18. The age-related features, including the recommended age-specific Mn dietary intake, age-specific water consumption rates, and age-specific homeostasis of Mn, are based on the available human data and knowledge of the biology of essential-metal homeostasis. Model simulations suggest that the impact of adding drinking-water exposure to daily Mn exposure via dietary intake and ambient air inhalation in children is not greater than the impacts in adults, even at a drinking-water concentration that is 2 times higher than the USEPA's lifetime health advisory value. This conclusion was also valid for formula-fed infants who are considered at the highest potential exposure to Mn from drinking water compared to all other age groups. Our multi-route, multi-source Mn PBPK model for infants and children provides insights about the potential for Mn-related health effects on growing children and will thereby improve the level of confidence in properly interpreting Mn exposure-health effects relationships in children in human epidemiological studies.


Subject(s)
Dietary Exposure/analysis , Drinking Water , Manganese/pharmacokinetics , Models, Biological , Water Pollutants, Chemical/pharmacokinetics , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant Formula , Infant, Newborn , Male , Milk, Human
4.
Regul Toxicol Pharmacol ; 70(1): 203-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25010378

ABSTRACT

ß-Chloroprene (2-chloro-1,3-butadiene, CD) is used in the manufacture of polychloroprene rubber. Chronic inhalation studies have demonstrated that CD is carcinogenic in B6C3F1 mice and Fischer 344 rats. However, epidemiological studies do not provide compelling evidence for an increased risk of mortality from total cancers of the lung. Differences between the responses observed in animals and humans may be related to differences in toxicokinetics, the metabolism and detoxification of potentially active metabolites, as well as species differences in sensitivity. The purpose of this study was to develop and apply a novel method that combines the results from available physiologically based kinetic (PBK) models for chloroprene with a statistical maximum likelihood approach to test commonality of low-dose risk across species. This method allows for the combined evaluation of human and animal cancer study results to evaluate the difference between predicted risks using both external and internal dose metrics. The method applied to mouse and human CD data supports the hypothesis that a PBK-based metric reconciles the differences in mouse and human low-dose risk estimates and further suggests that, after PBK metric exposure adjustment, humans are equally or less sensitive than mice to low levels of CD exposure.


Subject(s)
Carcinogens/toxicity , Chloroprene/toxicity , Neoplasms/chemically induced , Risk Assessment/methods , Animals , Carcinogens/administration & dosage , Carcinogens/pharmacokinetics , Chloroprene/administration & dosage , Chloroprene/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Likelihood Functions , Male , Mice , Neoplasms/epidemiology , Rats , Rats, Inbred F344 , Species Specificity
5.
Toxicol In Vitro ; 28(7): 1196-205, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24937311

ABSTRACT

In the recent National Research Council report on conducting a dose-response assessment for inorganic arsenic, the committee remarked that mode of action data should be used, to the extent possible, to extrapolate below the observed range for epidemiological studies to inform the shape of the dose-response curve. Recent in vitro mode of action studies focused on understanding the development of bladder cancer following exposure to inorganic arsenic provide data to inform the dose-response curve. These in vitro data, combined with results of bladder cancer epidemiology studies, inform the dose-response curve in the low-dose region, and include values for both pharmacokinetic and pharmacodynamic variability. Integration of these data provides evidence of a range of concentrations of arsenic for which no effect on the bladder would be expected. Specifically, integration of these results suggest that arsenic exposures in the range of 7-43 ppb in drinking water are exceedingly unlikely to elicit changes leading to key events in the development of cancer or noncancer effects in bladder tissue. These findings are consistent with the lack of evidence for bladder cancer following chronic ingestion of arsenic water concentrations <100 ppb in epidemiological studies.


Subject(s)
Arsenic/toxicity , Carcinogens/toxicity , Urinary Bladder Neoplasms/chemically induced , Water Pollutants, Chemical/toxicity , Animals , Arsenic/pharmacokinetics , Arsenic/standards , Carcinogens/pharmacokinetics , Carcinogens/standards , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Mice , Risk Assessment , Urinary Bladder Neoplasms/epidemiology , Urinary Bladder Neoplasms/metabolism , Water Pollutants, Chemical/pharmacokinetics , Water Pollutants, Chemical/standards
6.
Toxicol Sci ; 123(2): 421-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795629

ABSTRACT

Inorganic arsenic (As(i)) is a known human bladder carcinogen. The objective of this study was to examine the concentration dependence of the genomic response to As(i) in the urinary bladders of mice. C57BL/6J mice were exposed for 1 or 12 weeks to arsenate in drinking water at concentrations of 0.5, 2, 10, and 50 mg As/l. Urinary bladders were analyzed using gene expression microarrays. A consistent reversal was observed in the direction of gene expression change: from predominantly decreased expression at 1 week to predominantly increased expression at 12 weeks. These results are consistent with evidence from in vitro studies of an acute adaptive response that is suppressed on longer exposure due to downregulation of Fos. Pathways with the highest enrichment in gene expression changes were associated with epithelial-to-mesenchymal transition, inflammation, and proliferation. Benchmark dose (BMD) analysis determined that the lowest median BMD values for pathways were above 5 mg As/l, despite the fact that pathway enrichment was observed at the 0.5 mg As/l exposure concentration. This disparity may result from the nonmonotonic nature of the concentration-responses for the expression changes of a number of genes, as evidenced by the much fewer gene expression changes at 2 mg As/l compared with lower or higher concentrations. Pathway categories with concentration-related gene expression changes included cellular morphogenesis, inflammation, apoptosis/survival, cell cycle control, and DNA damage response. The results of this study provide evidence of a concentration-dependent transition in the mode of action for the subchronic effects of As(i) in mouse bladder cells in the vicinity of 2 mg As(i)/l.


Subject(s)
Arsenates/toxicity , Carcinogens, Environmental/toxicity , Epithelium/drug effects , Gene Expression/drug effects , Urinary Bladder/drug effects , Animals , Benchmarking , Dose-Response Relationship, Drug , Drinking , Epithelium/metabolism , Epithelium/pathology , Female , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Risk Assessment , Time Factors , Urinary Bladder/metabolism , Urinary Bladder/pathology , Water Supply
7.
Regul Toxicol Pharmacol ; 35(2 Pt 1): 177-97, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12052003

ABSTRACT

Increasing sophistication in methods used to account for human variability in susceptibility to toxicants has been one of the success stories in the continuing evolution of risk assessment science. Genetic polymorphisms have been suggested as an important contributor to overall human variability. Recently, data on polymorphisms in metabolic enzymes have been integrated with physiologically based pharmacokinetic (PBPK) modeling as an approach to determining the resulting overall variability. We present an analysis of the potential contribution of polymorphisms in enzymes modulating the disposition of four diverse compounds: methylene chloride, warfarin, parathion, and dichloroacetic acid. Through these case studies, we identify key uncertainties likely to be encountered in the use of polymorphism data and highlight potential simplifying assumptions that might be required to test the hypothesis that genetic factors are a substantive source of human variability in susceptibility to environmental toxicants. These uncertainties include (1) the relative contribution of multiple enzyme systems, (2) the extent of induction/inhibition through coexposure, (3) allelic frequencies of major ethnic groups, (4) the absence of chemical-specific data on the kinetic parameters for the different allelic forms of key enzymes, (5) large numbers of low-frequency alleles, and (6) uncertainty regarding differences between in vitro and in vivo kinetic data. Our effort sets the stage for the acquisition of critical data and further integration of polymorphism data with PBPK modeling as a means to quantitate population variability.


Subject(s)
Enzymes/genetics , Polymorphism, Genetic , Risk Assessment/methods , Xenobiotics/pharmacokinetics , Animals , Dichloroacetic Acid/pharmacokinetics , Dose-Response Relationship, Drug , Enzymes/metabolism , Humans , In Vitro Techniques , Methylene Chloride/pharmacokinetics , Parathion/pharmacokinetics , Reproducibility of Results , Uncertainty , Warfarin/pharmacokinetics
8.
Toxicol Sci ; 63(2): 160-72, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11568359

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model for isopropanol (IPA) and its major metabolite, acetone, is described. The structure of the parent chemical model, which can be used for either IPA or acetone by choosing the appropriate chemical-specific parameters, is similar to previously published models of volatile organic chemicals such as styrene. However, in order to properly simulate data on the exhalation of IPA and acetone during inhalation exposures, it was necessary to expand the description of the lung compartment to include a subcompartment for the upper respiratory tract mucus layer. This elaboration is consistent with published PBPK models of other water-soluble vapors in which the mucus layer serves to absorb the chemical during inhalation and then release it during exhalation. In the case of IPA exposure, a similar PBPK structure is used to describe the kinetics of the acetone produced from the metabolism of IPA. The resulting model is able to provide a coherent description of IPA and acetone kinetics in the rat and human for exposures to IPA by several routes: intravenous, intraperitoneal, oral, inhalation, and dermal. It is also able to consistently reproduce kinetic data for exposures of rats or humans to acetone. Thus, the model provides a validated framework for performing chemical-specific route-to-route extrapolation and cross-species dosimetry, which can be used in place of generic default calculations in support of risk assessments for IPA and acetone.


Subject(s)
2-Propanol/pharmacokinetics , Acetone/pharmacokinetics , Brain/drug effects , Metabolic Clearance Rate/drug effects , Models, Biological , 2-Propanol/metabolism , Absorption , Administration, Oral , Animals , Brain/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Humans , Inhalation Exposure , Kinetics , Liver/metabolism , Lung/metabolism , Mathematics , Permeability , Radiotherapy Planning, Computer-Assisted , Rats , Respiratory System/metabolism , Solubility , Tissue Distribution , Water
9.
Sci Total Environ ; 274(1-3): 37-66, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11453305

ABSTRACT

Vinyl chloride (VC) is a trans-species carcinogen, producing tumors in a variety of tissues, from both inhalation and oral exposures, across a number of species. In particular, exposure to VC has been associated with a rare tumor, liver angiosarcoma, in a large number of studies in mice, rats, and humans. The mode of action for the carcinogenicity of VC appears to be a relatively straightforward example of DNA adduct formation by a reactive metabolite, leading to mutation, mistranscription, and neoplasia. The objective of the present analysis was to investigate the comparative potency of a classic genotoxic carcinogen across species, by performing a quantitative comparison of the carcinogenic potency of VC using data from inhalation and oral rodent bioassays as well as from human epidemiological studies. A physiologically-based pharmacokinetic (PBPK) model for VC was developed to support the target tissue dosimetry for the cancer risk assessment. Unlike previous models, the initial metabolism of VC was described as occurring via two saturable pathways, one representing low capacity-high affinity oxidation by CYP2E1 and the other (in the rodent) representing higher capacity-lower affinity oxidation by other isozymes of P450, producing in both cases chloroethylene oxide (CEO) and chloroacetaldehyde (CAA) as intermediate reactive products. Depletion of glutathione by reaction with CEO and CAA was also described. Animal-based risk estimates for human inhalation exposure to VC using total metabolism estimates from the PBPK model were consistent with risk estimates based on human epidemiological data, and were lower than those currently used in environmental decision-making by a factor of 80.


Subject(s)
Carcinogens/toxicity , Neoplasms/chemically induced , Neoplasms/epidemiology , Vinyl Chloride/pharmacokinetics , Vinyl Chloride/toxicity , Air Pollutants/pharmacokinetics , Air Pollutants/toxicity , Animals , Carcinogens/pharmacokinetics , DNA Adducts , Humans , Mice , Models, Biological , Models, Statistical , Monte Carlo Method , Mutagenesis , Rats , Risk Assessment , Risk Factors , Transcription, Genetic
10.
Inhal Toxicol ; 13(5): 359-76, 2001 May.
Article in English | MEDLINE | ID: mdl-11295868

ABSTRACT

To assist in interspecies dosimetry comparisons for risk assessment of the nasal effects of organic acids, a hybrid computational fluid dynamics (CFD) and physiologically based pharmacokinetic (PBPK) dosimetry model was constructed to estimate the regional tissue dose of inhaled vapors in the rat and human nasal cavity. Application to a specific vapor would involve the incorporation of the chemical-specific reactivity, metabolism, partition coefficients, and diffusivity (in both air and tissue phases) of the vapor. This report describes the structure of the CFD-PBPK model and its application to a representative acidic vapor, acrylic acid, for interspecies tissue concentration comparisons to assist in risk assessment. By using the results from a series of short-term in vivo studies combined with computer modeling, regional nasal tissue dose estimates were developed and comparisons of tissue doses between species were conducted. To make these comparisons, the assumption was made that the susceptibilities of human and rat olfactory epithelium to the cytotoxic effects of organic acids were similar, based on similar histological structure and common mode of action considerations. Interspecies differences in response were therefore assumed to be driven primarily by differences in nasal tissue concentrations that result from regional differences in nasal air flow patterns relative to the species-specific distribution of olfactory epithelium in the nasal cavity. The results of simulations with the seven-compartment CFD-PBPK model suggested that the olfactory epithelium of the human nasal cavity would be exposed to tissue concentrations of acrylic acid similar to that of the rat nasal cavity when the exposure conditions are the same. Similar analysis of CFD data and CFD-PBPK model simulations with a simpler one-compartment model of the whole nasal cavities of rats and humans provides comparable results to averaging over the compartments of the seven-compartment model. These results indicate that the general structure of the hybrid CFD-PBPK model applied in this assessment would be useful for target tissue dosimetry and interspecies dose comparisons for a wide variety of vapors. Because of its flexibility, this CFD-PBPK model is envisioned to be a platform for the construction of case-specific inhalation dosimetry models to simulate in vivo exposures that do not involve significant histopathological damage to the nasal cavity.


Subject(s)
Acrylates/pharmacokinetics , Gases/pharmacokinetics , Inhalation Exposure/statistics & numerical data , Nasal Cavity/anatomy & histology , Acrylates/analysis , Acrylates/blood , Algorithms , Animals , Gases/analysis , Humans , Hydrogen-Ion Concentration , Models, Anatomic , Models, Biological , Nasal Cavity/metabolism , Nasal Mucosa/metabolism , Rats , Species Specificity , Tissue Distribution
11.
Environ Health Perspect ; 108 Suppl 2: 283-305, 2000 May.
Article in English | MEDLINE | ID: mdl-10807559

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model was developed that provides a comprehensive description of the kinetics of trichloroethylene (TCE) and its metabolites, trichloroethanol (TCOH), trichloroacetic acid (TCA), and dichloroacetic acid (DCA), in the mouse, rat, and human for both oral and inhalation exposure. The model includes descriptions of the three principal target tissues for cancer identified in animal bioassays: liver, lung, and kidney. Cancer dose metrics provided in the model include the area under the concentration curve (AUC) for TCA and DCA in the plasma, the peak concentration and AUC for chloral in the tracheobronchial region of the lung, and the production of a thioacetylating intermediate from dichlorovinylcysteine in the kidney. Additional dose metrics provided for noncancer risk assessment include the peak concentrations and AUCs for TCE and TCOH in the blood, as well as the total metabolism of TCE divided by the body weight. Sensitivity and uncertainty analyses were performed on the model to evaluate its suitability for use in a pharmacokinetic risk assessment for TCE. Model predictions of TCE, TCA, DCA, and TCOH concentrations in rodents and humans are in good agreement with a variety of experimental data, suggesting that the model should provide a useful basis for evaluating cross-species differences in pharmacokinetics for these chemicals. In the case of the lung and kidney target tissues, however, only limited data are available for establishing cross-species pharmacokinetics. As a result, PBPK model calculations of target tissue dose for lung and kidney should be used with caution.


Subject(s)
Carcinogens, Environmental/pharmacokinetics , Models, Biological , Risk Assessment , Trichloroethylene/pharmacokinetics , Animals , Carcinogens, Environmental/metabolism , Humans , Sensitivity and Specificity , Trichloroethylene/metabolism
12.
Toxicol Ind Health ; 16(9-10): 335-438, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11762928

ABSTRACT

Environmental risk-management decisions in the U.S. involving potential exposures to methylmercury currently use a reference dose (RfD) developed by the U.S. Environmental Protection Agency (USEPA). This RfD is based on retrospective studies of an acute poisoning incident in Iraq in which grain contaminated with a methylmercury fungicide was inadvertently used in the baking of bread. The exposures, which were relatively high but lasted only a few months, were associated with neurological effects in both adults (primarily paresthesia) and infants (late walking, late talking, etc.). It is generally believed that the developing fetus represents a particularly sensitive subpopulation for the neurological effects of methylmercury. The USEPA derived an RfD of 0.1 microg/kg/day based on benchmark dose (BMD) modeling of the combined neurological endpoints reported for children exposed in utero. This RfD included an uncertainty factor of 10 to consider human pharmacokinetic variability and database limitations (lack of data on multigeneration effects or possible long-term sequelae of perinatal exposure). Alcoa signed an Administrative Order of Consent for the conduct of a remedial investigation/feasibility study (RI/FS) at their Point Comfort Operations and the adjacent Lavaca Bay in Texas to address the effects of historical discharges of mercury-containing wastewater. In cooperation with the Texas Natural Resource Conservation Commission and USEPA Region VI, Alcoa conducted a baseline risk assessment to assess potential risk to human health and the environment. As a part of this assessment. Alcoa pursued the development of a site-specific RfD for methylmercury to specifically address the potential human health effects associated with the ingestion of contaminated finfish and shellfish from Lavaca Bay. Application of the published USEPA RfD to this site is problematic; while the study underlying the RfD represented acute exposure to relatively high concentrations of methylmercury, the exposures of concern for the Point Comfort site are from the chronic consumption of relatively low concentrations of methylmercury in fish. Since the publication of the USEPA RfD, several analyses of chronic exposure to methylmercury in fish-eating populations have been reported. The purpose of the analysis reported here was to evaluate the possibility of deriving an RfD for methylmercury, specifically for the case of fish ingestion, on the basis of these new studies. In order to better support the risk-management decisions associated with developing a remediation approach for the site in question, the analysis was designed to provide information on the distribution of acceptable ingestion rates across a population, which could reasonably be expected to be consistent with the results of the epidemiological studies of other fish-eating populations. Based on a review of the available literature on the effects of methylmercury, a study conducted with a population in the Seychelles Islands was selected as the critical study for this analysis. The exposures to methylmercury in this population result from chronic, multigenerational ingestion of contaminated fish. This prospective study was carefully conducted and analyzed, included a large cohort of mother-infant pairs, and was relatively free of confounding factors. The results of this study are essentially negative, and a no-observed-adverse-effect level (NOAEL) derived from the estimated exposures has recently been used by the Agency for Toxic Substances and Disease Registry (ATSDR) as the basis for a chronic oral minimal risk level (MRL) for methylmercury. In spite of the fact that no statistically significant effects were observed in this study, the data as reported are suitable for dose-response analysis using the BMD method. Evaluation of the BMD method used in this analysis, as well as in the current USEPA RfD, has demonstrated that the resulting 95% lower bound on the 10% benchmark dose (BMDL) represents a conservative estimate of the traditional NOAEL, and that it is superior to the use of "average" or "grouped" exposure estimates when dose-response information is available, as is the case for the Seychelles study. A more recent study in the Faroe Islands, which did report statistically significant associations between methylmercury exposure and neurological effects, could not be used for dose-response modeling due to inadequate reporting of the data and confounding from co-exposure to polychlorinated biphenyls (PCBs). BMD modeling over the wide range of neurological endpoints reported in the Seychelles study yielded a lowest BMDL for methylmercury in maternal hair of 21 ppm. This BMDL was then converted to an expected distribution of daily ingestion rates across a population using Monte Carlo analysis with a physiologically based pharmacokinetic (PBPK) model to evaluate the impact of interindividual variability. The resulting distribution of ingestion rates at the BMDL had a geometric mean of 1.60 microg/kg/day with a geometric standard deviation of 1.33; the 1st, 5th, and 10th percentiles of the distribution were 0.86, 1.04, and 1.15 microg/kg/day. In place of the use of an uncertainty factor of 3 for pharmacokinetic variability, as is done in the current RfD, one of these lower percentiles of the daily ingestion rate distribution provides a scientifically based, conservative basis for taking into consideration the impact of pharmacokinetic variability across the population. On the other hand, it was felt that an uncertainty factor of 3 for database limitations should be used in the current analysis. Although there can be high confidence in the benchmark-estimated NOAEL of 21 ppm in the Seychelles study, some results in the New Zealand and Faroe Islands studies could be construed to suggest the possibility of effects at maternal hair concentrations below 10 ppm. In addition, while concerns regarding the possibility of chronic sequelae are not supported by the available data, neither can they be absolutely ruled out. The use of an uncertainty factor of 3 is equivalent to using a NOAEL of 7 ppm in maternal hair, which provides additional protection against the possibility that effects could occur at lower concentrations in some populations. Based on the analysis described above, the distribution of acceptable daily ingestion rates (RfDs) recommended to serve as the basis for site-specific risk-management decisions at Alcoa's Point Comfort Operations ranges from approximately 0.3 to 1.1 microg/kg/day, with a population median (50th percentile) of 0.5 microg/kg/day. By analogy with USEPA guidelines for the use of percentiles in applications of distributions in exposure assessments, the 10th percentile provides a reasonably conservative measure. On this basis, a site-specific RfD of 0.4 microg/kg/day is recommended.


Subject(s)
Benchmarking , Environmental Exposure , Fishes , Food Contamination , Methylmercury Compounds/analysis , Models, Theoretical , Water Pollutants, Chemical/analysis , Adult , Animals , Cohort Studies , Female , Geography , Humans , Infant, Newborn , Male , Maternal-Fetal Exchange , Methylmercury Compounds/adverse effects , Methylmercury Compounds/pharmacokinetics , No-Observed-Adverse-Effect Level , Pregnancy , Public Health , Reference Values , Risk Assessment , Water Pollutants, Chemical/adverse effects , Water Pollutants, Chemical/pharmacokinetics
13.
Risk Anal ; 19(4): 547-58, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10765421

ABSTRACT

An analysis of the uncertainty in guidelines for the ingestion of methylmercury (MeHg) due to human pharmacokinetic variability was conducted using a physiologically based pharmacokinetic (PBPK) model that describes MeHg kinetics in the pregnant human and fetus. Two alternative derivations of an ingestion guideline for MeHg were considered: the U.S. Environmental Protection Agency reference dose (RfD) of 0.1 microgram/kg/day derived from studies of an Iraqi grain poisoning episode, and the Agency for Toxic Substances and Disease Registry chronic oral minimal risk level (MRL) of 0.5 microgram/kg/day based on studies of a fish-eating population in the Seychelles Islands. Calculation of an ingestion guideline for MeHg from either of these epidemiological studies requires calculation of a dose conversion factor (DCF) relating a hair mercury concentration to a chronic MeHg ingestion rate. To evaluate the uncertainty in this DCF across the population of U.S. women of child-bearing age, Monte Carlo analyses were performed in which distributions for each of the parameters in the PBPK model were randomly sampled 1000 times. The 1st and 5th percentiles of the resulting distribution of DCFs were a factor of 1.8 and 1.5 below the median, respectively. This estimate of variability is consistent with, but somewhat less than, previous analyses performed with empirical, one-compartment pharmacokinetic models. The use of a consistent factor in both guidelines of 1.5 for pharmacokinetic variability in the DCF, and keeping all other aspects of the derivations unchanged, would result in an RfD of 0.2 microgram/kg/day and an MRL of 0.3 microgram/kg/day.


Subject(s)
Methylmercury Compounds/administration & dosage , Methylmercury Compounds/pharmacokinetics , Adult , Female , Hair/metabolism , Humans , Male , Methylmercury Compounds/poisoning , Models, Biological , Monte Carlo Method , No-Observed-Adverse-Effect Level , Pregnancy , Risk Assessment
14.
J Toxicol Environ Health ; 52(6): 475-515, 1997 Dec 26.
Article in English | MEDLINE | ID: mdl-9397182

ABSTRACT

There has been relatively little attention given to incorporating knowledge of mode of action or of dosimetry of active toxic chemical to target tissue sites in the calculation of noncancer exposure guidelines. One exception is the focus in the revised reference concentration (RfC) process on delivered dose adjustments for inhaled materials. The studies reported here attempt to continue in the spirit of the new RfC guidelines by incorporating both mechanistic and delivered dose information using a physiologically based pharmacokinetic (PBPK) model, along with quantitative dose-response information using the benchmark dose (BMD) method, into the noncancer risk assessment paradigm. Two examples of the use of PBPK and BMD techniques in noncancer risk assessment are described: methylene chloride, and trichloroethylene. Minimal risk levels (MRLs) based on PBPK analysis of these chemicals were generally similar to those based on the traditional process, but individual MRLs ranged from roughly 10-fold higher to more than 10-fold lower than existing MRLs that were not based on PBPK modeling. Only two MRLs were based on critical studies that presented adequate data for BMD modeling, and in these two cases the BMD models were unable to provide an acceptable fit to the overall dose-response of the data, even using pharmacokinetic dose metrics. A review of 10 additional chemicals indicated that data reporting in the toxicological literature is often inadequate to support BMD modeling. Three general observations regarding the use of PBPK and BMD modeling in noncancer risk assessment were noted. First, a full PBPK model may not be necessary to support a more accurate risk assessment; often only a simple pharmacokinetic description, or an understanding of basic pharmacokinetic principles, is needed. Second, pharmacokinetic and mode of action considerations are a crucial factor in conducting noncancer risk assessments that involve animal-to-human extrapolation. Third, to support the application of BMD modeling in noncancer risk assessment, reporting of toxicity results in the toxicological literature should include both means and standard deviations for each dose group in the case of quantitative endpoints, such as relative organ weights or testing scores, and should report the number of animals affected in the case of qualitative endpoints.


Subject(s)
Models, Biological , Pharmacokinetics , Toxicology/methods , Animals , Dose-Response Relationship, Drug , Humans , Methylene Chloride/pharmacokinetics , Methylene Chloride/toxicity , Mice , Risk Assessment , Trichloroethylene/pharmacokinetics , Trichloroethylene/toxicity
15.
Chemosphere ; 31(1): 2561-78, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7670867

ABSTRACT

Risk assessments for vinyl chloride (VC) and trichloroethylene (TCE) are presented as examples of approaches for incorporating chemical-specific pharmacokinetic and mechanistic information into a more scientifically plausible cancer risk assessment. For VC, the evidence regarding mode of action includes direct reaction of a metabolite with DNA, resulting in DNA adducts and mistranscription, and cross-species target-tissue correspondence of a rare tumor type. Risk estimates for human exposure to VC predicted with a physiologically-based pharmacokinetic (PBPK) model and the linearized multistage (LMS) model were lower than those currently used in environmental decision-making by a factor of 30 to 50, and were more consistent with human epidemiological data. For TCE, there is evidence of increased cell proliferation due to receptor interaction or cytotoxicity in every instance in which tumors are observed, and the tumors typically represent an increase in the incidence of a commonly observed, species-specific lesion. Virtually safe exposure estimates for human exposure to TCE predicted with a PBPK model and a margin of exposure (MOE) approach were higher than those obtained by the conventional LMS approach by roughly a factor of 100. The MOE approach is recommended as an alternative to the LMS approach for chemicals with a carcinogenic mode of action which entails increased cell proliferation, leading to the expectation of a highly nonlinear cancer dose-response.


Subject(s)
Environmental Exposure , Environmental Pollutants/adverse effects , Neoplasms/chemically induced , Trichloroethylene/adverse effects , Vinyl Chloride/adverse effects , Animals , Cell Division/drug effects , DNA Adducts/metabolism , DNA Damage/drug effects , Dose-Response Relationship, Drug , Humans , Neoplasms/epidemiology , Risk Assessment , Transcription, Genetic/drug effects , Trichloroethylene/metabolism , Trichloroethylene/pharmacokinetics , Vinyl Chloride/metabolism , Vinyl Chloride/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...