Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Toxicol Sci ; 62(1): 124-39, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11399800

ABSTRACT

Methoxyethanol (ethylene glycol monomethyl ether, EGME), ethoxyethanol (ethylene glycol monoethyl ether, EGEE), and ethoxyethyl acetate (ethylene glycol monoethyl ether acetate, EGEEA) are all developmental toxicants in laboratory animals. Due to the imprecise nature of the exposure data in epidemiology studies of these chemicals, we relied on human and animal pharmacokinetic data, as well as animal toxicity data, to derive 3 occupational exposure limits (OELs). Physiologically based pharmacokinetic (PBPK) models for EGME, EGEE, and EGEEA in pregnant rats and humans have been developed (M. L. Gargas et al., 2000, Toxicol. Appl. Pharmacol. 165, 53-62; M. L. Gargas et al., 2000, Toxicol. Appl. Pharmacol. 165, 63-73). These models were used to calculate estimated human-equivalent no adverse effect levels (NAELs), based upon internal concentrations in rats exposed to no observed effect levels (NOELs) for developmental toxicity. Estimated NAEL values of 25 ppm for EGEEA and EGEE and 12 ppm for EGME were derived using average values for physiological, thermodynamic, and metabolic parameters in the PBPK model. The uncertainties in the point estimates for the NOELs and NAELs were estimated from the distribution of internal dose estimates obtained by varying key parameter values over expected ranges and probability distributions. Key parameters were identified through sensitivity analysis. Distributions of the values of these parameters were sampled using Monte Carlo techniques and appropriate dose metrics calculated for 1600 parameter sets. The 95th percentile values were used to calculate interindividual pharmacokinetic uncertainty factors (UFs) to account for variability among humans (UF(h,pk)). These values of 1.8 for EGEEA/EGEE and 1.7 for EGME are less than the default value of 3 for this area of uncertainty. The estimated human equivalent NAELs were divided by UF(h,pk) and the default UFs for pharmacodynamic variability among animals and among humans to calculate the proposed OELs. This methodology indicates that OELs (8-h time-weighted average) that should protect workers from the most sensitive adverse effects of these chemicals are 2 ppm EGEEA and EGEE (11 mg/m(3) EGEEA, 7 mg/m(3) EGEE) and 0.9 ppm (3 mg/m(3)) EGME. These recommendations assume that dermal exposure will be minimal or nonexistent.


Subject(s)
Ethylene Glycols/pharmacokinetics , Inhalation Exposure , Models, Biological , Monte Carlo Method , Occupational Exposure , Threshold Limit Values , Administration, Inhalation , Animals , Area Under Curve , Dose-Response Relationship, Drug , Ethylene Glycols/administration & dosage , Humans , No-Observed-Adverse-Effect Level , Species Specificity
2.
Toxicol Sci ; 60(2): 214-31, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11248133

ABSTRACT

Octamethylcyclotetrasiloxane (D4) is an ingredient in selected consumer and precision cleaning products. Workplace inhalation exposures may occur in some D4 production operations. In this study, we analyzed tissue, plasma, and excreta time-course data following D4 inhalation in Fischer 344 rats (K. Plotzke et al., 2000, Drug Metab. Dispos. 28, 192-204) to assess the degree to which the disposition of D4 is similar to or different from that of volatile hydrocarbons that lack silicone substitution. We first applied a basic physiologically based pharmacokinetic (PBPK) model (J. C. Ramsey and M. E. Andersen, 1984, Toxicol. Appl. Pharmacol. 73, 159-175) to characterize the biological determinants of D4 kinetics. Parameter estimation techniques indicated an unusual set of characteristics, i.e., a low blood:air (P(b:a) congruent with 0.9) and a high fat:blood partition coefficient (P(f:b) congruent with 550). These parameters were then determined experimentally by equilibrating tissue or liquid samples with saturated atmospheres of D4. Consistent with the estimates from the time-course data, blood:air partition coefficients were small, ranging from 1.9 to 6.9 in six samples. Perirenal fat:air partition coefficients were large, from 1400 to 2500. The average P(f:b) was determined to be 485. This combination of partitioning characteristics leads to rapid exhalation of free D4 at the cessation of the inhalation exposure followed by a much slower redistribution of D4 from fat and tissue storage compartments. The basic PK model failed to describe D4 tissue kinetics in the postexposure period and had to be expanded by adding deep-tissue compartments in liver and lung, a mobile chylomicron-like lipid transport pool in blood, and a second fat compartment. Model parameters for the refined model were optimized using single-exposure data in male and female rats exposed at three concentrations: 7, 70, and 700 ppm. With inclusion of induction of D4 metabolism at 700 ppm (3-fold in males, 1-fold in females), the parameter set from the single exposures successfully predicted PK results from 14-day multiple exposures at 7 and 700 ppm. A common parameter set worked for both genders. Despite its very high lipophilicity, D4 does not show prolonged retention because of high hepatic and exhalation clearance. The high lipid solubility, low blood:air partition coefficient, and plasma lipid storage with D4 led to novel distributional characteristics not previously noted for inhaled organic hydrocarbons. These novel characteristics were only made apparent by analysis of the time-course data with PBPK modeling techniques.


Subject(s)
Models, Biological , Siloxanes/pharmacokinetics , Administration, Inhalation , Animals , Female , Inhalation Exposure , Male , Rats , Rats, Inbred F344 , Siloxanes/administration & dosage , Tissue Distribution
3.
Drug Metab Dispos ; 28(2): 192-204, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10640518

ABSTRACT

The retention, distribution, metabolism, and excretion of [(14)C]octamethylcyclotetrasiloxane (D(4)) were studied in Fischer 344 rats after single and multiple exposures to 7, 70, or 700 ppm [(14)C]D(4). Subset groups were established for body burden, distribution, and elimination. Retention of inhaled D(4) was relatively low (5-6% of inhaled D(4)). Radioactivity derived from [(14)C]D(4) inhalation was widely distributed to tissues of the rat. Maximum concentrations of radioactivity in plasma and tissues (except fat) occurred at the end of exposure and up to 3 h postexposure. Maximum concentrations of radioactivity in fat occurred as late as 24 h postexposure. Fat was a depot, elimination of radioactivity from this tissue was much slower than from plasma and other tissues. With minor exceptions, there were no consistent gender effects on the distribution of radioactivity and the concentrations of radioactivity were nearly proportional to exposure concentration over the exposure range. Excretion of radioactivity was via exhaled breath and urine, and, to a much lesser extent, feces. Urinary metabolites included dimethylsilanediol and methylsilanetriol plus five minor metabolites. Relative abundance of these metabolites was the same from every test group. Elimination was rapid during the first 24 h after exposure and was slower thereafter (measured up to 168 h postexposure). In singly-exposed female (but not male) rats, small dose-dependent shifts in elimination pathways were seen. After multiple exposures, the elimination pathways were dose- and gender-independent. These data define possible pathways for metabolism of D(4) and allow estimation of the persistence of D(4) and/or its metabolites in rats.


Subject(s)
Siloxanes/pharmacokinetics , Administration, Inhalation , Animals , Area Under Curve , Atmosphere Exposure Chambers , Body Burden , Female , Half-Life , Male , Rats , Rats, Inbred F344 , Respiratory Function Tests , Sex Characteristics , Siloxanes/administration & dosage , Tissue Distribution
4.
Toxicology ; 113(1-3): 231-7, 1996 Oct 28.
Article in English | MEDLINE | ID: mdl-8901903

ABSTRACT

The National Academy of Sciences and many others have noted the need for quantitative health risk assessment methodology that goes beyond a simple screening analysis based on upper bounds on risk. The Academy recommended adoption of methodologies which provide a higher-tier analysis based on realistic estimates of risk which reflect more of the available biological information. In recent years, scientists have challenged the assumption of low-dose linearity and other default assumptions in cancer risk assessment. These challenges have stimulated the continued evolution of quantitative risk assessment methodologies, because effective risk management requires accurate characterizations of uncertainty and greater utilization of cost-benefit analyses for decision making. "Comprehensive Realism" is an emerging quantitative weight-of-evidence based risk assessment methodology for both cancer and noncancer health effects which utilizes probability distributions and decision analysis techniques to reflect more of the available human and animal dose-response data. The current state of knowledge about the relative plausibility of alternative dose-response analyses is also addressed in this approach. The framework discussed here should lead to a higher-tier assessment of butadiene.


Subject(s)
Butadienes/toxicity , Carcinogens/toxicity , Risk Assessment , Butadienes/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Models, Biological
5.
Toxicol Appl Pharmacol ; 137(2): 253-67, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8661351

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model capable of describing the metabolism of vinyl chloride (VC) in rats, mice, and humans has been developed and validated by comparison with experimental data from experiments not used in model development. This PBPK model has been used to predict measures of delivered dose (reactive VC metabolites produced in the livers of the affected species) hypothesized to be involved in the induction of liver angiosarcoma in rats, mice, and human populations exposed to VC. Measures of delivered dose in rats were fit to an empirical dose-response model (the linearized multistage model of Crump et al.) and used to make predictions of liver angiosarcoma incidence in mice and human populations exposed to VC. This procedure gave a good prediction of angiosarcoma incidence in mice. Predictions of angiosarcoma incidence in humans were more than two orders of magnitude lower than risk estimations which did not utilize pharmacokinetic data, but were still almost an order of magnitude higher than actually observed in exposed human populations.


Subject(s)
Carcinogens/pharmacokinetics , Carcinogens/toxicity , Models, Biological , Neoplasms/chemically induced , Vinyl Chloride/pharmacokinetics , Vinyl Chloride/toxicity , Animals , Carcinogens/metabolism , Female , Hemangiosarcoma/chemically induced , Hemangiosarcoma/epidemiology , Humans , Incidence , Liver Neoplasms/chemically induced , Liver Neoplasms/epidemiology , Male , Mice , Mice, Inbred Strains , Neoplasms, Experimental/chemically induced , Predictive Value of Tests , Rabbits , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Risk Assessment , Stimulation, Chemical , Vinyl Chloride/metabolism
6.
Toxicol Appl Pharmacol ; 136(2): 289-306, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8619237

ABSTRACT

In vivo experiments in rats and mice and in vitro experiments in rats, mice, and humans have been used to develop and validate a "2nd generation" physiologically based pharmacokinetic (PBPK) model for perchloroethylene (PERC). The refined PBPK model should be useful in the preparation of carcinogenic risk assessment based on amounts of PERC metabolites formed in the livers of rodents and humans according to procedures developed by EPA. A sensitivity analysis of the PBPK model revealed that the most significant uncertainties in this process (other than the choice of the appropriate dose/response model based on mechanism of action of PERC) were in the techniques used to estimate rates of PERC metabolism in humans. In vitro studies with human tissues reported help define what some have called the range of "equally reasonable alternatives" for estimating human risk.


Subject(s)
Models, Biological , Solvents/metabolism , Tetrachloroethylene/metabolism , Administration, Inhalation , Animals , Biotransformation , Computer Simulation , Dose-Response Relationship, Drug , Gases/pharmacokinetics , Humans , In Vitro Techniques , Male , Mice , Microsomes, Liver/metabolism , Rats , Rats, Inbred F344 , Reproducibility of Results , Risk Assessment , Sensitivity and Specificity , Solvents/administration & dosage , Solvents/pharmacokinetics , Solvents/toxicity , Tetrachloroethylene/administration & dosage , Tetrachloroethylene/pharmacokinetics , Tetrachloroethylene/toxicity
7.
Toxicol Lett ; 64-65 Spec No: 669-76, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1471221

ABSTRACT

Because of the expense involved in conducting chronic studies, limited numbers of animals and dose groups are used. This has given rise to the practice of including as one of the dose groups the "Maximum Tolerated Dose" (MTD). This dose is operationally defined as the highest dose which can be administered to animals without adversely affecting their survival through effects other than cancer. Since many detoxification systems in animals are capacity-limited, they frequently become saturated in MTD studies. This may lead to difficulties in interpreting the results of MTD studies, particularly when it is necessary to estimate the hazard for human populations whose exposure is typically much lower than the MTD. For this reason, it is important to characterize the dose-dependency of absorption, distribution, metabolism, and elimination (pharmacokinetics) of test substances prior to the initiation of a chronic study. This provides a basis for determining the number and spacing of doses to be used in a chronic study. If the appropriate information is collected it may also be possible to develop a physiologically based pharmacokinetic model which facilitates extrapolation of the toxicity results between different species and routes of administration as well as between high and low doses. For instance, methylene chloride and vinyl chloride are predominantly metabolized by saturable oxidative pathway(s) at low exposure concentrations. In each case, the oxidative pathway saturates at exposures much lower than the MTD. Knowledge of the pharmacokinetic behavior of these substances provided a basis for appropriately interpreting the chronic studies which have been conducted with these materials.


Subject(s)
Biphenyl Compounds/toxicity , Methylene Chloride/toxicity , Vinyl Chloride/toxicity , Animals , Biotransformation , Biphenyl Compounds/metabolism , Dose-Response Relationship, Drug , Methylene Chloride/metabolism , Rats , Vinyl Chloride/metabolism
8.
Toxicol Appl Pharmacol ; 108(1): 14-27, 1991 Mar 15.
Article in English | MEDLINE | ID: mdl-1900959

ABSTRACT

Dichloromethane (methylene chloride, DCM) and other dihalomethanes are metabolized to carbon monoxide (CO) which reversibly binds hemoglobin and is eliminated by exhalation. We have developed a physiologically based pharmacokinetic (PB-PK) model which describes the kinetics of CO, carboxyhemoglobin (HbCO), and parent dihalomethane, and have applied this model to examine the inhalation kinetics of CO and of DCM in rats and humans. The portion of the model describing CO and HbCO kinetics was adapted from the Coburn-Forster-Kane equation, after modification to include production of CO by DCM oxidation. DCM kinetics and metabolism were described by a generic PB-PK model for volatile chemicals (RAMSEY AND ANDERSEN, Toxicol. Appl. Pharmacol. 73, 159-175, 1984). Physiological and biochemical constants for CO were first estimated by exposing rats to 200 ppm CO for 2 hr and examining the time course of HbCO after cessation of CO exposure. These CO inhalation studies provided estimates of CO diffusing capacity under free breathing and for the Haldane coefficient, the relative equilibrium distribution ratio for hemoglobin between CO and O2. The CO model was then coupled to a PB-PK model for DCM to predict HbCO time course behavior during and after DCM exposures in rats. By coupling the models it was possible to estimate the yield of CO from oxidation of DCM. In rats only about 0.7 mol of CO are produced from 1 mol of DCM during oxidation. The combined model adequately represented HbCO and DCM behavior following 4-hr exposures to 200 or 1000 ppm DCM, and HbCO behavior following 1/2-hr exposure to 5160 ppm DCM or 5000 ppm bromochloromethane. The rat PB-PK model was scaled to predict DCM, HbCO, and CO kinetics in humans exposed either to DCM or to CO. Three human data sets from the literature were examined: (1) inhalation of CO at 50, 100, 250, and 500 ppm; (2) seven 1/2-hr inhalation exposures to 50, 100, 250, and 500 ppm DCM; and (3) 2-hr inhalation exposures to 986 ppm DCM. An additional data set from human volunteers exposed to 100 or 350 ppm DCM for 6 hr is reported here for the first time. Endogenous CO production rates and the initial amount of CO in the blood compartment were varied in each study as necessary to give the baseline HbCO value, which varied from less than 0.5% to greater than 2% HbCO. The combined PB-PK model gave a good representation of the observed behavior in all four human studies.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Carbon Dioxide/metabolism , Carboxyhemoglobin/metabolism , Methylene Chloride/pharmacokinetics , Models, Biological , Administration, Inhalation , Animals , Humans , Male , Rats , Rats, Inbred F344
9.
Ann Ist Super Sanita ; 27(4): 609-14, 1991.
Article in English | MEDLINE | ID: mdl-1820733

ABSTRACT

Dichloromethane (methylene chloride, CH2Cl2) has been shown to significantly increase the incidence of malignant lung and liver tumors in B6C3F1 mice inhaling high concentrations of CH2Cl2 vapor for the majority of their natural lifetime. CH2Cl2 is extensively metabolized in mammalian species through two competing pathways: (1) oxidation by the mixed function oxidase enzymes, and (2) conjugation with glutathione catalyzed by glutathione-S-transferase(s)(GST). Since elevated tumor incidences have not been observed in B6C3F1 mice exposed to 1,1,1-trichloroethane, a halogenated solvent with similar physical-chemical properties (but only minor amounts of mammalian metabolism), it appeared that biologically reactive intermediates (BRIs) from one or both of the pathways of CH2Cl2 metabolism were involved in the tumorigenic process. Development of an integrated pharmacokinetic model incorporating quantitative measures of mammalian physiology, chemical solubility, and metabolic rate constants permitted formulation of a plausible hypothesis for the tumorigenic effects of CH2Cl2: namely that BRIs formed by the CH2Cl2/GST(s) may react with critical molecules in the target organs. This hypothesis is consistent with the dose-dependency, route-dependency, and species-specificity of CH2Cl2 for the induction of lung and liver tumors. Based on this hypothesis as well as in vivo and in vitro measurements of CH2Cl2 metabolism in humans, it was possible to prepare quantitative estimates of the cancer risk in human populations. Examination of these risk estimates indicates that development of quantitative procedures for describing the production of BRI in target tissues may cause significant changes in the levels of estimated risk.


Subject(s)
Carcinogenicity Tests , Liver Neoplasms, Experimental/chemically induced , Lung Neoplasms/chemically induced , Methylene Chloride/pharmacokinetics , Administration, Inhalation , Animals , Biotransformation , Glutathione Transferase/metabolism , Humans , Methylene Chloride/administration & dosage , Mice , Mice, Inbred Strains , Mixed Function Oxygenases/metabolism , Models, Biological , Risk Factors , Species Specificity
10.
Adv Exp Med Biol ; 283: 649-55, 1991.
Article in English | MEDLINE | ID: mdl-2069033

ABSTRACT

Development of quantitative mathematical models for the production of BRIs in target tissues can provide valuable insights into the mechanisms of toxicity for specific chemicals. Furthermore, use of mechanistic information and mathematical modeling in the hazard evaluation process should significantly reduce the uncertainty inherent in extrapolating the results of animal toxicity tests to man.


Subject(s)
Methylene Chloride/toxicity , Neoplasms/chemically induced , Animals , Glutathione/metabolism , Humans , Methylene Chloride/administration & dosage , Methylene Chloride/metabolism , Mixed Function Oxygenases/metabolism , Models, Biological
11.
Toxicol Appl Pharmacol ; 105(3): 443-59, 1990 Sep 15.
Article in English | MEDLINE | ID: mdl-2237918

ABSTRACT

A physiologically based pharmacokinetic (PB-PK) model for CHCl3 has been used to prepare estimates of the probability that human populations exposed to low levels of CHCl3 will develop liver tumors similar to those seen in rodent bioassays. The PB-PK model for CHCl3 was based on a model reported earlier by Corley et al. (1990), but this model differed from that of Corley et al. in that it was also capable of describing a pharmacodynamic endpoint: induction of cytotoxicity in the liver of CHCl3-exposed animals produced by reactive metabolites of CHCl3. Pharmacodynamic descriptions in this model were derived from experimental measurements of cell replication ([3H]thymidine incorporation) as well as from quantitative histopathology in the liver of rats and mice. Two different approaches were used for hazard evaluation: (1) a "Safety Factor" approach based on no observed effect levels for liver tumors. and (2) calculation of lower confidence limits on risk-specific doses with the GLOBAL83 computer program. In each case, cytotoxicity produced by reactive CHCl3 metabolites was used as the measure of "dose" to the liver. The Safety Factor approach suggested that continuous exposure of human populations to concentrations of CHCl3 less than 2840 ppb in air or 13,900 ppb in water would not be likely to significantly increase the risk of developing liver tumors. The second approach suggested a "plausible upper 95% confidence limit" of 1 x 10(-5) for lifetime excess cancer risk for human populations continuously exposed to 2200 or 13,100 ppb CHCl3 in air or water, respectively.


Subject(s)
Chloroform/toxicity , Liver Neoplasms/chemically induced , Administration, Inhalation , Administration, Oral , Animals , Cell Division , Cell Survival/drug effects , Chloroform/administration & dosage , Chloroform/metabolism , Chloroform/pharmacokinetics , Computer Simulation , Dose-Response Relationship, Drug , Female , Humans , Macromolecular Substances , Male , Mice , Models, Biological , Proportional Hazards Models , Rats , Risk Factors , Software
12.
Toxicol Appl Pharmacol ; 105(1): 37-54, 1990 Aug.
Article in English | MEDLINE | ID: mdl-2392805

ABSTRACT

A six compartment physiologically based pharmacokinetic (PB-PK) model was developed to describe the disposition of diethylene-1,4-dioxide (dioxane) and its principal metabolite beta-hydroxyethoxyacetic acid in rats, mice, and humans. The model was developed from experimentally measured partition coefficients (reported here for the first time) as well as pharmacokinetic data previously reported. The completed PB-PK model adequately described data from gavage and intravenous studies in rats, as well as inhalation studies in rats and humans. Substantial nonlinearities were observed in the kinetic behavior of dioxane under high exposure conditions (water concentrations greater than 0.1% dioxane and atmospheric concentrations greater than 300 ppm dioxane). The PB-PK model was subsequently used to prepare quantitative estimates of the "plausible upper bounds" on carcinogenic risk for human populations exposed to dioxane in air or water. Based on these quantitative estimates, it appears that human populations continuously exposed to 740-3700 ppb dioxane in air or 20,000-120,000 ppb dioxane in water would be unlikely to experience increased frequencies of tumors.


Subject(s)
Carcinogens, Environmental/pharmacokinetics , Dioxanes/pharmacokinetics , Dioxins/pharmacokinetics , Models, Biological , Animals , Computer Simulation , Dioxanes/administration & dosage , Dioxanes/toxicity , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Rats , Risk , Solubility
13.
Toxicol Appl Pharmacol ; 103(3): 512-27, 1990 May.
Article in English | MEDLINE | ID: mdl-2339423

ABSTRACT

A physiologically based pharmacokinetic model describing the disposition of chloroform in mice, rats, and humans was developed. This model was designed to facilitate extrapolations from high doses, such as those used in chronic rodent studies, to low doses that humans may be exposed to in the workplace or the environment. Kinetic constants for mice and rats were derived from in vivo experiments. Enzymatic studies conducted with samples of rodent and human tissues provided a rational basis for estimating human in vivo metabolic rate constants. Incorporation of physiological descriptions of the processes of absorption, distribution, metabolism, and excretion allowed extrapolation between different routes of exposure as well. The model was validated by comparing model predictions with experimental data gathered in mice, rats, and humans after inhalation, oral, or intraperitoneal administration of chloroform. Consistent with previous reports, the metabolic activation of chloroform to toxic intermediates was shown to occur most rapidly in the mouse, less rapidly in the rat, and most slowly in humans. Estimates of the "delivered dose" of chloroform metabolites to internal organs sensitive to chloroform toxicity were calculated. This model may be used to develop refined dose estimates for human populations exposed to low levels of chloroform in the environment.


Subject(s)
Chloroform/pharmacokinetics , Models, Biological , Administration, Inhalation , Administration, Oral , Animals , Carbon Radioisotopes , Chloroform/administration & dosage , Chloroform/metabolism , Cricetinae , Drinking , Evaluation Studies as Topic , Humans , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Rats , Rats, Inbred Strains , Water
14.
Toxicol Appl Pharmacol ; 97(2): 230-46, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2922756

ABSTRACT

Physiologically based pharmacokinetic (PB-PK) models describe the dynamic behavior of chemicals and their metabolites in individual tissues of living animals. Because PB-PK models contain specific parameters related to the physiological and biochemical properties of different species as well as the physical chemical characteristics of individual chemicals, they are useful tools for performing high dose/low dose, dose route, and interspecies extrapolations in hazard evaluations. An example of such extrapolation has been presented by M. E. Andersen, H. J. Clewell III, M. L. Gargas, F. A. Smith, and R. H. Reitz (Toxicol. Appl. Pharmacol. 87, 185-205, 1987), who employed a PB-PK model for methylene chloride (CH2Cl2) to estimate the chronic toxicity of this material. However, one limitation of this PB-PK model was that the metabolic rate constants for the glutathione-S-transferase (GST) pathway in humans were estimated by allometric scaling rather than from experimental data. In this paper we report studies designed to estimate the in vivo rates of metabolism of CH2Cl2 from in vitro incubations of lung and liver tissues from B6C3F1 mice, F344 rats, Syrian Golden hamsters, and humans. A procedure for calculating in vivo metabolic rate constants from the in vitro studies is presented. This procedure was validated by making extrapolations with mixed function oxidase enzymes (MFO) acting on CH2Cl2, where both in vitro and in vivo rates of metabolism are known. The in vitro rate constants for the two enzyme systems are consistent with the hypothesis presented by Andersen et al. that metabolism of CH2Cl2 occurs in vivo by two competing pathways: a high-affinity saturable pathway (identified as MFO) and a low-affinity first-order pathway (identified as GST). The metabolic rate constants for GST obtained from these studies are also consistent with the hypothesis of Andersen et al. that production of large quantities of glutathione/CH2Cl2 conjugates in vivo may increase the frequency with which lung and liver tumors develop in some species of animals (e.g., B6C3F1 mouse). When in vivo studies in humans are unavailable, in vitro enzyme assays provide a reasonable method for estimating metabolic rate constants.


Subject(s)
Hydrocarbons, Chlorinated/pharmacokinetics , Methylene Chloride/pharmacokinetics , Animals , Cricetinae , Glutathione/analysis , Humans , In Vitro Techniques , Kinetics , Liver/enzymology , Lung/enzymology , Male , Methylene Chloride/toxicity , Mice , Mice, Inbred Strains , Mixed Function Oxygenases/analysis , Models, Biological , Rats , Rats, Inbred F344 , Risk Factors
15.
Regul Toxicol Pharmacol ; 8(4): 408-13, 1988 Dec.
Article in English | MEDLINE | ID: mdl-3222482

ABSTRACT

Toxicokinetics provides a powerful tool, which is not used sufficiently in the conduct and interpretation of animal toxicity studies. In selecting doses for toxicity studies toxicokinetic data can be used effectively. The tools of toxicokinetics are limited only by the toxicologists' understanding of basic biologic mechanisms. More knowledge on mechanisms of action implicates the possibility of more detailed toxicokinetic models. Physiologically based pharmacokinetic models are valuable in the interpretation of animal toxicity studies. They provide a physiological basis for extrapolating between species and routes of administration, and the future use of these models in setting acceptable levels of exposure for non-cancer toxicity in humans deserves serious consideration.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Pharmacokinetics , Animals , Data Interpretation, Statistical , Research Design
16.
Toxicol Lett ; 43(1-3): 189-200, 1988 Oct.
Article in English | MEDLINE | ID: mdl-3176064

ABSTRACT

Chemical carcinogenesis is a process beginning with carcinogen absorption and ending with development of a malignant tumor. Individual elements of this process have been studied intensively but no comprehensive model has been developed. This report describes a comprehensive model which incorporates carcinogen pharmacokinetics, biochemical mechanism of action, and the resultant mutation of normal cells to malignancy. Model parameters correspond to specific physiological and biochemical structures and processes. The model was encoded in a simulation language and used to examined biochemical and cellular effects of exposure to an initiator and a promoter. With laboratory validation, the model should be useful for interpretation and design of studies on carcinogenic mechanisms and for risk assessment.


Subject(s)
Carcinogens/pharmacokinetics , Computer Simulation , Mutation , Neoplasms/chemically induced , DNA Damage , DNA-Binding Proteins/physiology , Probability
17.
Toxicol Lett ; 43(1-3): 97-116, 1988 Oct.
Article in English | MEDLINE | ID: mdl-3176073

ABSTRACT

Physiologically-based pharmacokinetic (PB-PK) models provide a mechanism for reducing the uncertainty inherent in extrapolating the results of animal toxicity tests to man. This paper discusses a technique for incorporating data from in vitro studies of xenobiotic metabolism into in vivo PB-PK models. Methylene chloride is used as an example, and carcinogenic risk estimates incorporating PB-PK principles are presented.


Subject(s)
Hydrocarbons, Chlorinated/toxicity , Methylene Chloride/toxicity , Models, Biological , Animals , Cricetinae , Cytosol/enzymology , Glutathione Transferase/metabolism , Humans , In Vitro Techniques , Liver/enzymology , Lung/enzymology , Methylene Chloride/pharmacokinetics , Mice , Microsomes/enzymology , Mixed Function Oxygenases/metabolism , Neoplasms, Experimental/chemically induced , Rats , Risk Factors , Xenobiotics/toxicity
18.
Toxicol Appl Pharmacol ; 95(2): 185-99, 1988 Sep 15.
Article in English | MEDLINE | ID: mdl-3420611

ABSTRACT

A unified physiologically based pharmacokinetic (PB-PK) model was developed and used to describe the disposition of methylchloroform (1,1,1-trichloroethane, MC) in three different species (rats, mice, and humans) after four different routes of exposure (inhalation, intravenous injection, bolus gavage, and drinking water administration). Metabolism of MC followed Michaelis-Menten kinetics in each species. Vmax's were calculated from the allometric equation: Vmax = 0.419 BW0.7, and Km appeared to be identical in each species (5.75 mg equivalents/liter). Once the PB-PK model had been developed for young adult animals (1-3 months of age), it was used to study the disposition of MC in older rats and mice (approximately 18.5 months of age). Most of the changes in the pharmacokinetic behavior of MC in older rats could be simulated by increasing the size of the fat compartment in the PB-PK model from 7 to 18% of body weight. However, the pharmacokinetic behavior in older mice was more complex; increasing the size of the fat compartment in this species from 4 to 18% only accounted for part of the observed differences between old and young animals. An appropriate dose surrogate (average area under the liver concentration/time curve) was selected and the PB-PK model was used to make quantitative comparisons between "internal doses" of MC in long term animal studies and "internal doses" associated with human exposures to MC. Values of the dose surrogate in humans consuming 2 liters/day of water with typical levels of MC contamination (1-10 ppb) were four to six orders of magnitude lower than the dose surrogates in the rodent studies at levels of MC exposure which failed to produce adverse effects on the liver (875-1500 ppm, 6 hr/day, 5 days/week).


Subject(s)
Hydrocarbons, Chlorinated/pharmacokinetics , Trichloroethanes/pharmacokinetics , Animals , Breath Tests , Drinking , Humans , Male , Mice , Models, Biological , Rats , Rats, Inbred F344 , Trichloroethanes/administration & dosage
20.
J Toxicol Sci ; 12(2): 223-33, 1987 May.
Article in English | MEDLINE | ID: mdl-3625818

ABSTRACT

While much of industrial toxicology is observational in character, pursuit of specific research is needed to facilitate the overall evaluation of potential toxicity for man. Two such areas are the application of physiologic pharmacokinetic models to inter-species extrapolation of toxic effects and an understanding of the role of cellular oncogenes in the process of spontaneous tumor formation in animals. A physiologic pharmacokinetic model was developed for methylene chloride (MeCl2) which describes the fate of MeCl2 and its metabolic products in numerous species including the mouse, rat, hamster and man. This model has been used to predict specific tissue concentrations of critical metabolic reaction products in target tissues between animals and man. If it is assumed that toxicity is related to target tissue concentrations such methodology provides a means of relating interspecies toxicity to absorbed dose. This methodology precludes the necessity of using arbitrary factors in relating animal toxicity data to man. A particular controversial issue in animal toxicology is the significance of the enhancement of animal tumors in tissues which already have a high spontaneous incidence. Without a better understanding of the basic process of spontaneous tumor formation it remains difficult to interpret results from chemical treatment. In particular spontaneous liver tumors in the B6C3F1 mouse have been shown to contain an activated cellular oncogene identified as H-RAS. The activated cellular oncogene is present in tumor tissue only and not in surrounding normal liver tissue. Of particular significance is the high frequency of activation in these mouse liver tumors (82%) compared to a 10-20% incidence of oncogenes present in a variety of human tumors. This suggests the ultra sensitivity of this mouse strain to liver tumor induction. Additional studies in progress are designed to determine whether genotoxic and nongenotoxic hepatocarcinogens show differences in oncogene activation.


Subject(s)
Toxicology/methods , Animals , Dose-Response Relationship, Drug , Humans , Kinetics , Liver Neoplasms, Experimental/etiology , Methylene Chloride/metabolism , Models, Biological , Oncogenes , Species Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL