Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Toxicol Environ Health A ; 75(21): 1280-97, 2012.
Article in English | MEDLINE | ID: mdl-23030654

ABSTRACT

A screening-level risk assessment was conducted for styrene-acrylonitrile (SAN) Trimer detected at the Reich Farm Superfund site in Toms River, NJ. Consistent with a screening-level approach, on-site and off-site exposure scenarios were evaluated using assumptions that are expected to overestimate actual exposures and hazards at the site. Environmental sampling data collected for soil and groundwater were used to estimate exposure point concentrations. Several exposure scenarios were evaluated to assess potential on-site and off-site exposures, using parameter values for exposures to soil (oral, inhalation of particulates, and dermal contact) and groundwater (oral, dermal contact) to reflect central tendency exposure (CTE) and reasonable maximum exposure (RME) conditions. Three reference dose (RfD) values were derived for SAN Trimer for short-term, subchronic, and chronic exposures, based upon its effects on the liver in exposed rats. Benchmark (BMD) methods were used to assess the relationship between exposure and response, and to characterize appropriate points of departure (POD) for each RfD. An uncertainty factor of 300 was applied to each POD to yield RfD values of 0.1, 0.04, and 0.03 mg/kg-d for short-term, subchronic, and chronic exposures, respectively. Because a chronic cancer bioassay for SAN Trimer in rats (NTP 2011a) does not provide evidence of carcinogenicity, a cancer risk assessment is not appropriate for this chemical. Potential health hazards to human health were assessed using a hazard index (HI) approach, which considers the ratio of exposure dose (i.e., average daily dose, mg/kg-d) to toxicity dose (RfD, mg/kg-d) for each scenario. All CTE and RME HI values are well below 1 (where the average daily dose is equivalent to the RfD), indicating that there is no concern for potential noncancer effects in exposed populations even under the conservative assumptions of this screening-level assessment.


Subject(s)
Acrylonitrile/analysis , Groundwater/analysis , Soil Pollutants/analysis , Styrene/analysis , Water Pollutants, Chemical/analysis , Acrylonitrile/toxicity , Animals , Environmental Exposure/analysis , Environmental Monitoring/methods , Humans , Rats , Risk Assessment , Soil Pollutants/toxicity , Styrene/toxicity , Water Pollutants, Chemical/toxicity
2.
Inhal Toxicol ; 21(6): 462-79, 2009 May.
Article in English | MEDLINE | ID: mdl-19519147

ABSTRACT

Methyl iodide (MeI) induces fetotoxicity in New Zealand White (NZW) rabbits when maternal exposure occurs during a susceptible window late in gestation (gestation days [GD] 23-26). To identify the possible mode of action, comprehensive maternal and fetal bioanalysis and thyroid structure/function assessments were conducted in MeI-exposed (25 ppm by whole-body inhalation) and unexposed time-mated NZW rabbits (10/group) during GD 21-27. Key developmental events were observed within this window in unexposed fetuses, including the appearance of colloid in the thyroid follicular lumen and the detection of serum T(3) beginning on GD 22. Fetal T(4) and T(3) levels were diminished following maternal MeI exposure compared to baseline values. Fetal TSH was significantly increased following 4 days of maternal MeI exposure. MeI-induced changes in the fetal thyroid included reduced colloid formation, epithelial follicular hypertrophy, and epithelial cytoplasmic vacuolation. Time-course investigations using 20 ppm MeI revealed highly concentrated levels of iodide in fetal versus maternal serum. Direct maternal administration of sodium iodide by intravenous infusion during GD 23-26 induced similar effects on fetal thyroid structure and function as MeI, identifying iodide as the putative agent. Elevated S-methylcysteine adduct concentrations were noted in fetal hemoglobin, indicating that some unreacted MeI may be delivered directly to the fetus. However, the weight of evidence from these studies suggests that late-stage fetal death following maternal exposure to MeI during GD 23-26 is the result of preferential accumulation of iodide in the fetal compartment causing disruption of the fetal hypothalamic-pituitary-thyroid axis at a critical time in the development of the rabbit fetal thyroid.


Subject(s)
Fetal Death/chemically induced , Hydrocarbons, Iodinated/toxicity , Hypothyroidism/chemically induced , Maternal Exposure/adverse effects , Animals , Female , Fetal Death/blood , Gestational Age , Hydrocarbons, Iodinated/blood , Hypothyroidism/blood , Inhalation Exposure/adverse effects , Pregnancy , Rabbits
3.
Inhal Toxicol ; 21(6): 512-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19519151

ABSTRACT

The percentages of total airflows over the nasal respiratory and olfactory epithelium of female rabbits were calculated from computational fluid dynamics (CFD) simulations of steady-state inhalation. These airflow calculations, along with nasal airway geometry determinations, are critical parameters for hybrid CFD/physiologically based pharmacokinetic models that describe the nasal dosimetry of water-soluble or reactive gases and vapors in rabbits. CFD simulations were based upon three-dimensional computational meshes derived from magnetic resonance images of three adult female New Zealand White (NZW) rabbits. In the anterior portion of the nose, the maxillary turbinates of rabbits are considerably more complex than comparable regions in rats, mice, monkeys, or humans. This leads to a greater surface area to volume ratio in this region and thus the potential for increased extraction of water soluble or reactive gases and vapors in the anterior portion of the nose compared to many other species. Although there was considerable interanimal variability in the fine structures of the nasal turbinates and airflows in the anterior portions of the nose, there was remarkable consistency between rabbits in the percentage of total inspired airflows that reached the ethmoid turbinate region (approximately 50%) that is presumably lined with olfactory epithelium. These latter results (airflows reaching the ethmoid turbinate region) were higher than previous published estimates for the male F344 rat (19%) and human (7%). These differences in regional airflows can have significant implications in interspecies extrapolations of nasal dosimetry.


Subject(s)
Magnetic Resonance Imaging/methods , Models, Biological , Nasal Cavity/physiology , Pulmonary Ventilation/physiology , Animals , Computational Biology/methods , Computer Simulation , Female , Inhalation Exposure/adverse effects , Inhalation Exposure/standards , Magnetic Resonance Imaging/standards , Maximal Expiratory Flow Rate/physiology , Nasal Cavity/anatomy & histology , Rabbits
4.
Food Chem Toxicol ; 47(4): 760-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19166901

ABSTRACT

Acrylamide in cooked foods results in wide-spread, low-level human exposure. Potential risks from dietary intake remain unclear due to apparent conflicting results from cancer bioassays conducted in rats that reported tumors and epidemiology studies that are suggestive but provide little or no evidence of increased cancer. Risk estimation often includes two common assumptions: (1) tumor response rates in test species can be extrapolated systematically to estimate human response rates and (2) tumor rates observed following high-dose exposures can be linearly extrapolated to predict response rates following low-dose exposures. The validity of these assumptions was evaluated for acrylamide based upon the examination of relevant toxicokinetic and toxicodynamic differences between humans and rats, including sources of nonlinearity that modify high to low dose extrapolation of cancer incidence. Important species differences and sources of nonlinearity are identified, and recommendations for addressing them within the quantitative framework of a PBTK/TD model are discussed. These differences are likely to estimate risk levels up to several orders of magnitude lower in humans than in rats. Quantitative inclusion of these TK/TD factors will more closely estimate actual human cancer risk derived from high-dose rodent studies, since detoxification processes for acrylamide and glycidamide appear adequately protective against toxicity from human dietary doses.


Subject(s)
Acrylamide/toxicity , Risk Assessment , Acrylamide/pharmacokinetics , Animals , Blood Proteins/metabolism , Cytochrome P-450 Enzyme System/physiology , DNA Damage , Dose-Response Relationship, Drug , Epoxide Hydrolases/physiology , Humans , Models, Biological , Protein Binding , Species Specificity
5.
Risk Anal ; 28(5): 1375-94, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18761732

ABSTRACT

Dose-response assessments were conducted for the noncancer effects of acrylonitrile (AN) for the purposes of deriving subchronic and chronic oral reference dose (RfD) and inhalation reference concentration (RfC) values. Based upon an evaluation of available toxicity data, the irritation and neurological effects of AN were determined to be appropriate bases for deriving reference values. A PBPK model, which describes the toxicokinetics of AN and its metabolite 2-cyanoethylene oxide (CEO) in both rats and humans, was used to assess the dose-response data in terms of an internal dose measure for the oral RfD values, but could not be used in deriving the inhalation RfC values. Benchmark dose (BMD) methods were used to derive all reference values. Where sufficient information was available, data-derived uncertainty factors were applied to the points of departure determined by BMD methods. From this assessment, subchronic and chronic oral RfD values of 0.5 and 0.05 mg/kg/day, respectively, were derived. Similarly, subchronic and chronic inhalation RfC values of 0.1 and 0.06 mg/m(3), respectively, were derived. Confidence in the reference values derived for AN was considered to be medium to high, based upon a consideration of the confidence in the key studies, the toxicity database, dosimetry, and dose-response modeling.


Subject(s)
Acrylonitrile/administration & dosage , Carcinogens/administration & dosage , Acrylonitrile/pharmacokinetics , Acrylonitrile/toxicity , Administration, Inhalation , Administration, Oral , Animal Experimentation , Animals , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Clinical Trials as Topic , Dose-Response Relationship, Drug , Humans , Rats , Reference Values
6.
J Toxicol Environ Health A ; 70(21): 1838-48, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17934956

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model was developed for inhaled ethylbenzene (EB) in B6C3F1 mice. The mouse physiological parameters were obtained from the literature, but the blood:air and tissue:air partition coefficients were determined by vial equilibration technique. The maximal velocity for hepatic metabolism (Vmax) obtained from a previously published rat study was increased by a factor of approximately 3 to account for enzyme induction during repeated exposures. The Michaelis affinity constant (Km) for hepatic metabolism of EB, obtained from a previously published rat PBPK modeling study, was kept unchanged during single and repeated exposure scenarios. Hepatic metabolism alone could not adequately describe the clearance of EB from mouse blood. Additional metabolism was assumed to be localized in the lung. The parameters for pulmonary metabolism were obtained by optimization of PBPK model fits to kinetic data collected following exposures to 75-1000 ppm. The PBPK model successfully predicted all available blood and tissue concentration data in mice exposed to 75 or 750 ppm EB. Overall, the results indicate that the rate of EB clearance is markedly higher in B6C3F1 mice than rats or humans and exceeds the hepatic metabolism capacity. Available biochemical evidence is consistent with a significant role for pulmonary metabolism; however, the extent to which the extrahepatic metabolism is localized in the lung is unclear. Overall, the PBPK model developed for the mouse adequately simulated the blood and tissue kinetics of EB by accounting for its high rate of clearance.


Subject(s)
Benzene Derivatives/pharmacokinetics , Liver/metabolism , Lung/metabolism , Models, Biological , Administration, Inhalation , Animals , Benzene Derivatives/administration & dosage , Benzene Derivatives/blood , Cardiac Output , Female , Male , Metabolic Clearance Rate , Mice
7.
Regul Toxicol Pharmacol ; 43(1): 85-103, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16099568

ABSTRACT

A cancer dose-response assessment was conducted for acrylonitrile (AN) using updated information on mechanism of action, epidemiology, toxicity, and pharmacokinetics. Although more than 10 chronic bioassays indicate that AN produces multiple tumors in rats and mice, a number of large, well-conducted epidemiology studies provide no evidence of a causal association between AN exposure and cancer mortality of any type. The epidemiological data include early industry exposures that are far higher than occur today and that approach or exceed levels found to be tumorigenic in animals. Despite the absence of positive findings in the epidemiology data, a dose-response assessment was conducted for AN based on brain tumors in rats. Mechanistic studies implicate the involvement of oxidative stress in rat brain due to a metabolite (2-cyanoethylene oxide or CEO, cyanide), but do not conclusively rule out a potential role for the direct genotoxicity of CEO. A PBPK model was used to predict internal doses (peak CEO in brain) for 12 data sets, which were pooled together to provide a consistent characterization of the dose-response relationship for brain tumor incidence in the rat. The internal dose corresponding to a 5% increase in extra risk (ED 05=0.017 mg/L brain) and its lower confidence limit (LED 05=0.014 mg/L brain) was used as the point of departure. The weight-of-evidence supports the use of a nonlinear extrapolation for the cancer dose-response assessment. A quantitative comparison of the epidemiology exposure-response data (lung and brain cancer mortality) to the rat brain tumor data in terms of internal dose adds to the confidence in the nonlinear extrapolation. Uncertainty factors of 200 and 220 (for the oral and inhalation routes, respectively) were applied to the LED 05 to account for interspecies variation, intraspecies variation, and the severity of the response measure. Accordingly, oral doses below 0.009 mg/kg-day and air concentrations below 0.1mg/m(3) are not expected to pose an appreciable risk to human populations exposed to AN.


Subject(s)
Acrylonitrile/toxicity , Brain Neoplasms/chemically induced , Carcinogens/toxicity , Environmental Exposure/standards , Lung Neoplasms/chemically induced , Acrylonitrile/administration & dosage , Administration, Oral , Animals , Brain Neoplasms/epidemiology , Brain Neoplasms/mortality , Carcinogenicity Tests , Carcinogens/administration & dosage , Dose-Response Relationship, Drug , Humans , Incidence , Inhalation Exposure , Lung Neoplasms/epidemiology , Lung Neoplasms/mortality , Meta-Analysis as Topic , Mice , Models, Biological , Mutagenicity Tests , Nonlinear Dynamics , Rats
8.
Risk Anal ; 25(2): 271-84, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15876203

ABSTRACT

Reference values, including an oral reference dose (RfD) and an inhalation reference concentration (RfC), were derived for propylene glycol methyl ether (PGME), and an oral RfD was derived for its acetate (PGMEA). These values were based on transient sedation observed in F344 rats and B6C3F1 mice during a two-year inhalation study. The dose-response relationship for sedation was characterized using internal dose measures as predicted by a physiologically-based pharmacokinetic (PBPK) model for PGME and its acetate. PBPK modeling was used to account for changes in rodent physiology and metabolism due to aging and adaptation, based on data collected during Weeks 1, 2, 26, 52, and 78 of a chronic inhalation study. The peak concentration of PGME in richly perfused tissues (i.e., brain) was selected as the most appropriate internal dose measure based on a consideration of the mode of action for sedation and similarities in tissue partitioning between brain and other richly perfused tissues. Internal doses (peak tissue concentrations of PGME) were designated as either no-observed-adverse-effect levels (NOAELs) or lowest-observed-adverse-effect levels (LOAELs) based on the presence or the absence of sedation at each time point, species, and sex in the two-year study. Distributions of the NOAEL and LOAEL values expressed in terms of internal dose were characterized using an arithmetic mean and standard deviation, with the mean internal NOAEL serving as the basis for the reference values, which was then divided by appropriate uncertainty factors. Where data were permitting, chemical-specific adjustment factors were derived to replace default uncertainty factor values of 10. Nonlinear kinetics, which was predicted by the model in all species at PGME concentrations exceeding 100 ppm, complicate interspecies, and low-dose extrapolations. To address this complication, reference values were derived using two approaches that differ with respect to the order in which these extrapolations were performed: (1) default approach of interspecies extrapolation to determine the human equivalent concentration (PBPK modeling) followed by uncertainty factor application, and (2) uncertainty factor application followed by interspecies extrapolation (PBPK modeling). The resulting reference values for these two approaches are substantially different, with values from the latter approach being seven-fold higher than those from the former approach. Such a striking difference between the two approaches reveals an underlying issue that has received little attention in the literature regarding the application of uncertainty factors and interspecies extrapolations to compounds where saturable kinetics occur in the range of the NOAEL. Until such discussions have taken place, reference values based on the former approach are recommended for risk assessments involving human exposures to PGME and PGMEA.


Subject(s)
Propylene Glycols/chemistry , Propylene Glycols/pharmacokinetics , Administration, Inhalation , Administration, Oral , Age Factors , Aging , Animals , Dose-Response Relationship, Drug , Female , Hypnotics and Sedatives/pharmacology , Kinetics , Male , Mice , Models, Chemical , Models, Theoretical , Nonlinear Dynamics , Perfusion , Rabbits , Rats , Rats, Inbred F344 , Reference Standards , Risk Assessment , Sex Factors , Time Factors
9.
Risk Anal ; 24(5): 1165-83, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15563286

ABSTRACT

Ethylene oxide (EO) has been identified as a carcinogen in laboratory animals. Although the precise mechanism of action is not known, tumors in animals exposed to EO are presumed to result from its genotoxicity. The overall weight of evidence for carcinogenicity from a large body of epidemiological data in the published literature remains limited. There is some evidence for an association between EO exposure and lympho/hematopoietic cancer mortality. Of these cancers, the evidence provided by two large cohorts with the longest follow-up is most consistent for leukemia. Together with what is known about human leukemia and EO at the molecular level, there is a body of evidence that supports a plausible mode of action for EO as a potential leukemogen. Based on a consideration of the mode of action, the events leading from EO exposure to the development of leukemia (and therefore risk) are expected to be proportional to the square of the dose. In support of this hypothesis, a quadratic dose-response model provided the best overall fit to the epidemiology data in the range of observation. Cancer dose-response assessments based on human and animal data are presented using three different assumptions for extrapolating to low doses: (1) risk is linearly proportionate to dose; (2) there is no appreciable risk at low doses (margin-of-exposure or reference dose approach); and (3) risk below the point of departure continues to be proportionate to the square of the dose. The weight of evidence for EO supports the use of a nonlinear assessment. Therefore, exposures to concentrations below 37 microg/m3 are not likely to pose an appreciable risk of leukemia in human populations. However, if quantitative estimates of risk at low doses are desired and the mode of action for EO is considered, these risks are best quantified using the quadratic estimates of cancer potency, which are approximately 3.2- to 32-fold lower, using alternative points of departure, than the linear estimates of cancer potency for EO. An approach is described for linking the selection of an appropriate point of departure to the confidence in the proposed mode of action. Despite high confidence in the proposed mode of action, a small linear component for the dose-response relationship at low concentrations cannot be ruled out conclusively. Accordingly, a unit risk value of 4.5 x 10(-8) (microg/m3)(-1) was derived for EO, with a range of unit risk values of 1.4 x 10(-8) to 1.4 x 10(-7) (microg/m3)(-1) reflecting the uncertainty associated with a theoretical linear term at low concentrations.


Subject(s)
Carcinogens/toxicity , Ethylene Oxide/toxicity , Mutagens/toxicity , Neoplasms/chemically induced , Animals , Carcinogens/administration & dosage , Dose-Response Relationship, Drug , Ethylene Oxide/administration & dosage , Female , Humans , Leukemia/chemically induced , Male , Mice , Models, Biological , Mutagens/administration & dosage , Nonlinear Dynamics , Rats , Risk Assessment
10.
Regul Toxicol Pharmacol ; 38(3): 345-67, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14623485

ABSTRACT

The performance of allometric scaling of dose as a power of body weight under a variety of extrapolation conditions with respect to species, route, exposure intensity, and mechanism/mode of action, remains untested in many cases. In this paper, animal-human internal dose ratio comparisons have been developed for 12 chemicals (benzene, carbon tetrachloride, chloroform, diisopropylfluorophosphate, ethanol, ethylene oxide, methylene chloride, methylmercury, styrene, tetrachloroethene, trichloroethene, and vinyl chloride). This group of predominantly volatile and lipophilic chemicals was selected on the basis that their kinetics have been well-studied and can be predicted in mice, rats, and humans using physiologically based pharmacokinetic (PBPK) models. PBPK model predictions were compared to the allometric scaling predictions for interspecies extrapolation. Recommendations for the application of the allometric scaling are made with reference to internal dose measure (mode of action) and concentration level. The results of this assessment generally support the use of scaling factors recommended in the published literature, which includes scaling factors of 1.0 for risk assessments in which toxicity is attributed to the parent chemical or stable metabolite, and -0.75 for dose-response assessments in which toxicity is attributed to the formation of a reactive metabolite from an inhaled compound. A scaling factor of 0.75 is recommended for dose-response assessments of orally administered compounds in which toxicity is attributed to the parent chemical or stable metabolite and 1.0 for risk assessments in which toxicity is attributed to the formation of a reactive metabolite from a compound administered by the oral route. A dose-dependency in the results suggests that the scaling factors appropriate at high exposures may differ from those at low exposures, primarily due to the impact of saturable metabolism.


Subject(s)
Anthropometry/methods , Dose-Response Relationship, Drug , Inactivation, Metabolic/physiology , Risk Assessment/methods , Administration, Inhalation , Administration, Oral , Animals , Benzene/administration & dosage , Benzene/pharmacokinetics , Drug Evaluation, Preclinical , Ethanol/administration & dosage , Ethanol/pharmacokinetics , Ethylene Oxide/administration & dosage , Ethylene Oxide/pharmacokinetics , Humans , Hydrocarbons, Chlorinated/administration & dosage , Hydrocarbons, Chlorinated/pharmacokinetics , Isoflurophate/administration & dosage , Isoflurophate/pharmacokinetics , Methylmercury Compounds/administration & dosage , Methylmercury Compounds/pharmacokinetics , Mice , Models, Biological , Rats , Styrene/administration & dosage , Styrene/pharmacokinetics
11.
Environ Sci Technol ; 35(14): 2917-25, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11478243

ABSTRACT

Bisphenol A (BPA), a high-volume chemical used to make polycarbonate plastic, epoxy resins, and other chemicals has been reported to be weakly estrogenic. To investigate the effects of long-term exposure to Bisphenol A, a multigeneration study was conducted in which fathead minnows (Pimephales promelas) were exposed to water concentrations of BPA in the range from 1 to 1280 micrograms/L. In this paper, we report the growth and reproductive effects of BPA on sexually mature adults in the F0 generation (after 43, 71, and 164 d of exposure) and the effects on hatchability in the F1 generation. Mean measured concentrations of BPA in the water for all doses, over a 164-d exposure period, were between 70% and 96% of nominal. An inhibitory effect of BPA on somatic growth (length and weight) occurred in adult male fish exposed to 640 and 1280 micrograms/L (after 71 and 164 d). BPA induced vitellogenin synthesis (VTG; a biomarker for estrogen exposure) in males at concentrations of 640 and 1280 micrograms/L after 43 d and 160 micrograms/L after 71 d. In females, plasma VTG concentrations were elevated above controls only after 164-d exposure to 640 micrograms/L. Inhibition of gonadal growth (as measured by the gonadosomatic index) occurred in both males and females at concentrations of 640 and 1280 micrograms/L after 164 d. In males, a concentration of 16 micrograms/L altered the proportion of sex cell types in the testis, suggesting inhibition of spermatogenesis. Concentrations of BPA that induced VTG synthesis and affected gonadal development were lower than those that resulted in discernible effects on reproductive output. Egg production was inhibited at a BPA concentration of 1280 micrograms/L, and hatchability in the F1 generation was reduced at a BPA concentration of 640 micrograms/L (there were not enough eggs spawned in the 1280 micrograms/L group for hatchability studies to be conducted). The results demonstrate that BPA acts as a weak estrogen to fish when administered via the water, with effects on breeding at and above 640 micrograms/L.


Subject(s)
Cyprinidae/physiology , Estrogens, Non-Steroidal/adverse effects , Phenols/adverse effects , Reproduction/drug effects , Water Pollutants, Chemical/adverse effects , Animals , Benzhydryl Compounds , Dose-Response Relationship, Drug , Environmental Exposure , Female , Gonads/drug effects , Gonads/growth & development , Male , Vitellogenins/biosynthesis , Vitellogenins/blood
12.
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
13.
Chem Biol Interact ; 135-136: 303-22, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11397398

ABSTRACT

Potential health effects of human exposure to 1,3-butadiene (BD) are of concern due to the use of BD in industry and its low-level presence throughout the environment. Physiologically based toxicokinetic (PBTK) models of BD in rodents have been developed by multiple research groups in an effort to explain species differences in toxicity (especially carcinogenic potency) through toxicokinetics. PBTK modeling of dose metrics related to a non-cancer endpoint, ovotoxicity in experimental animals, was conducted. The cumulative area under the blood concentration vs. time curve (AUC) for the metabolite diepoxybutane (butadiene diepoxide, DEB) was found to be consistent with ovotoxicity in mice and rats exposed to BD by inhalation or epoxybutene (butadiene monoepoxide, EB) or DEB by intraperitoneal injection. This suggests that cumulative DEB AUC may also be an appropriate metric for possible human risk. A preliminary human PBTK model was assembled for the eventual assessment of reproductive risk to humans and for prioritizing the determination of model parameters. The preliminary model accurately predicted published data on exhaled breath BD concentrations in a human volunteer exposed to BD by inhalation. The fit was relatively insensitive to the rate constant for BD epoxidation. Sensitivity analyses were conducted on this human PBTK model. Using a range of published rate constants, human blood DEB was found to be sensitive to rates of epoxidation of EB to DEB and hydrolysis of EB and DEB, but not BD epoxidation. Because of the large ranges of rates measured in vitro for these reactions, different combinations of in-vitro rates produce varying predictions of blood DEB concentration. Thus, validation of a human PBTK model with human biomonitoring data will be essential to produce a PBTK model that can be applied to risk assessment.


Subject(s)
Butadienes/metabolism , Butadienes/toxicity , Models, Biological , Animals , Epoxy Compounds/metabolism , Female , Humans , Mice , Rats , Risk Assessment , Sensitivity and Specificity , Species Specificity
14.
Risk Anal ; 20(1): 135-51, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10795346

ABSTRACT

Historically, U.S. regulators have derived cancer slope factors by using applied dose and tumor response data from a single key bioassay or by averaging the cancer slope factors of several key bioassays. Recent changes in U.S. Environmental Protection Agency (EPA) guidelines for cancer risk assessment have acknowledged the value of better use of mechanistic data and better dose-response characterization. However, agency guidelines may benefit from additional considerations presented in this paper. An exploratory study was conducted by using rat brain tumor data for acrylonitrile (AN) to investigate the use of physiologically based pharmacokinetic (PBPK) modeling along with pooling of dose-response data across routes of exposure as a means for improving carcinogen risk assessment methods. In this study, two contrasting assessments were conducted for AN-induced brain tumors in the rat on the basis of (1) the EPA's approach, the dose-response relationship was characterized by using administered dose/concentration for each of the key studies assessed individually; and (2) an analysis of the pooled data, the dose-response relationship was characterized by using PBPK-derived internal dose measures for a combined database of ten bioassays. The cancer potencies predicted for AN by the contrasting assessments are remarkably different (i.e., risk-specific doses differ by as much as two to four orders of magnitude), with the pooled data assessments yielding lower values. This result suggests that current carcinogen risk assessment practices overestimate AN cancer potency. This methodology should be equally applicable to other data-rich chemicals in identifying (1) a useful dose measure, (2) an appropriate dose-response model, (3) an acceptable point of departure, and (4) an appropriate method of extrapolation from the range of observation to the range of prediction when a chemical's mode of action remains uncertain.


Subject(s)
Acrylonitrile/adverse effects , Brain Neoplasms/diagnosis , Carcinogens/adverse effects , Models, Biological , Acrylonitrile/administration & dosage , Acrylonitrile/pharmacokinetics , Air Pollutants/adverse effects , Air Pollutants/pharmacokinetics , Animals , Biological Assay , Brain/metabolism , Carcinogens/administration & dosage , Carcinogens/pharmacokinetics , Databases as Topic , Dose-Response Relationship, Drug , Environmental Exposure , Ethylene Oxide/adverse effects , Ethylene Oxide/analogs & derivatives , Ethylene Oxide/pharmacokinetics , Forecasting , Guidelines as Topic , Linear Models , Nonlinear Dynamics , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Risk Assessment , United States , United States Environmental Protection Agency , Water Pollutants, Chemical/administration & dosage , Water Pollutants, Chemical/adverse effects , Water Pollutants, Chemical/pharmacokinetics
15.
Toxicol Appl Pharmacol ; 165(1): 1-26, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10814549

ABSTRACT

Ethylene (ET) is a gaseous olefin of considerable industrial importance. It is also ubiquitous in the environment and is produced in plants, mammals, and humans. Uptake of exogenous ET occurs via inhalation. ET is biotransformed to ethylene oxide (EO), which is also an important volatile industrial chemical. This epoxide forms hydroxyethyl adducts with macromolecules such as hemoglobin and DNA and is mutagenic in vivo and in vitro and carcinogenic in experimental animals. It is metabolically eliminated by epoxide hydrolase and glutathione S-transferase and a small fraction is exhaled unchanged. To estimate the body burden of EO in rodents and human resulting from exposures to EO and ET, we developed a physiological toxicokinetic model. It describes uptake of ET and EO following inhalation and intraperitoneal administration, endogenous production of ET, enzyme-mediated oxidation of ET to EO, bioavailability of EO, EO metabolism, and formation of 2-hydroxyethyl adducts of hemoglobin and DNA. The model includes compartments representing arterial, venous, and pulmonary blood, liver, muscle, fat, and richly perfused tissues. Partition coefficients and metabolic parameters were derived from experimental data or published values. Model simulations were compared with a series of data collected in rodents or humans. The model describes well the uptake, elimination, and endogenous production of ET in all three species. Simulations of EO concentrations in blood and exhaled air of rodents and humans exposed to EO or ET were in good agreement with measured data. Using published rate constants for the formation of 2-hydroxyethyl adducts with hemoglobin and DNA, adduct levels were predicted and compared with values reported. In humans, predicted hemoglobin adducts resulting from exposure to EO or ET are in agreement with measured values. In rodents, simulated and measured DNA adduct levels agreed generally well, but hemoglobin adducts were underpredicted by a factor of 2 to 3. Obviously, there are inconsistencies between measured DNA and hemoglobin adduct levels.


Subject(s)
DNA Adducts , Ethylene Oxide/pharmacokinetics , Ethylenes/pharmacokinetics , Hemoglobins/metabolism , Animals , Disinfectants/metabolism , Disinfectants/pharmacokinetics , Disinfectants/toxicity , Ethylene Oxide/metabolism , Ethylene Oxide/toxicity , Ethylenes/metabolism , Ethylenes/toxicity , Humans , Inhalation Exposure , Kinetics , Metabolic Clearance Rate , Mice , Models, Biological , Rats , Rats, Sprague-Dawley , Tissue Distribution
16.
Toxicol Appl Pharmacol ; 165(1): 53-62, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10814553

ABSTRACT

Exposures to sufficiently high doses of ethylene glycol monomethyl ether (2-methoxyethanol, 2-ME) have been found to produce developmental effects in rodents and nonhuman primates. The acetic acid metabolite of 2-ME, 2-methoxyacetic acid (2-MAA), is the likely toxicant, and, as such, an understanding of the kinetics of 2-MAA is important when assessing the potential risks to humans associated with 2-ME. A previously described physiologically based pharmacokinetic (PBPK) model of 2-ME/2-MAA kinetics for rats exposed via oral or iv administration was extended and validated to inhalation exposures. Pregnant Sprague-Dawley rats were exposed for 5 days (gestation days 11-15), 6 h/day, to 2-ME vapor at 10 and 50 ppm. Validation consisted of comparing model output to maternal blood and fetal 2-ME and 2-MAA concentrations during and following 5 days of exposure (gestation days 11-15). These concentrations correspond to a known no observed effect level (NOEL) and a lowest observed effect level (LOEL) for developmental effects in rats. The rat PBPK model for 2-ME/2-MAA was scaled to humans and the model (without the pregnancy component) was used to predict data collected by other investigators on the kinetics of 2-MAA excretion in urine following exposures to 2-ME in human volunteers. The partially validated human model (with the pregnancy component) was used to predict equivalent human exposure concentrations based on 2-MAA dose measures (maximum blood concentration, C(max), and average daily area under the 2-MAA blood concentration curve, AUC, during pregnancy) that correspond to the concentrations measured at the rat NOEL and LOEL exposure concentrations. Using traditional PBPK scale-up techniques, it was calculated that pregnant women exposed for 8 h/day, 5 days/week, for the duration of pregnancy would need to be exposed to 12 or 60 ppm 2-ME to produce maternal 2-MAA blood concentrations (C(max) or average daily AUC) equivalent to those in rats exposed to the NOEL (10 ppm) or LOEL (50 ppm), respectively.


Subject(s)
Ethylene Glycols/pharmacokinetics , Teratogens/pharmacokinetics , Acetates/pharmacokinetics , Acetates/urine , Animals , Ethylene Glycols/toxicity , Ethylene Glycols/urine , Female , Humans , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/urine , Inhalation Exposure , Models, Biological , Predictive Value of Tests , Pregnancy , Radiotherapy Planning, Computer-Assisted , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Teratogens/toxicity
17.
Toxicol Appl Pharmacol ; 165(1): 63-73, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10814554

ABSTRACT

The solvents ethylene glycol monoethyl ether acetate (EGEEA) and ethylene glycol monoethyl ether (EGEE), at sufficiently high doses, are known to be rodent developmental toxicants, exerting their toxic effects through the action of their metabolite 2-ethoxyacetic acid (2-EAA). Thus risks associated with exposure to these compounds are best evaluated based on a measure of the internal dose of 2-EAA. The goals of the work reported here were to develop physiologically based pharmacokinetic (PBPK) models of EGEEA and EGEE for pregnant rats and humans. These models were used to identify human exposure levels (ppm in air) equivalent to the rat no observed effect level (NOEL) and lowest observed effect level (LOEL) for developmental effects (Hanley et al., 1984). We exposed pregnant Sprague-Dawley rats to concentrations of EGEEA corresponding to the NOEL and LOEL. Maternal blood, urine, and fetal tissue concentrations of EGEE and 2-EAA measured in these experiments were used to validate the rat EGEEA and EGEE models. Data collected by other researchers were used to validate the capabilities of the rodent EGEEA and EGEE models to predict the kinetics in humans. The models for estimating circulating blood concentrations of 2-EAA were considered valid based on the ability of the model to accurately predict 2-EAA concentrations in rat blood, urine, and fetal tissue. The human inhaled concentration equivalent to the rat NOEL for EGEEA (50 ppm) was predicted to be 25 ppm using the maternal blood average daily area under the curve (AUC) and 40 ppm using the maximum concentration achieved in maternal blood (C(max)). The human inhaled concentration equivalent to the rat LOEL for EGEEA (100 ppm) was determined to be 55 ppm using the maternal blood average daily AUC and 80 ppm using the maternal blood C(max).


Subject(s)
Ethylene Glycols/pharmacokinetics , Teratogens/pharmacokinetics , Animals , Environmental Exposure , Ethylene Glycols/toxicity , Female , Humans , Inhalation Exposure , Models, Biological , Occupational Exposure , Predictive Value of Tests , Pregnancy , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Teratogens/toxicity
18.
Toxicol Appl Pharmacol ; 163(1): 67-74, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10662606

ABSTRACT

An accurate description of developing embryos' exposure to a xenobiotic is a desirable component of mechanism-based risk assessments for humans exposed to potential developmental toxicants during pregnancy. 2-Methoxyethanol (2-ME), a solvent used in the manufacture of semiconductors, is embryotoxic and teratogenic in all species tested including nonhuman primates. 2-Methoxyacetic acid (2-MAA) is the primary metabolite of 2-ME and the proximate embryotoxic agent. The objective of the work described here was to adapt an existing physiologically based pharmacokinetic (PBPK) model for 2-ME and 2-MAA kinetics during midorganogenesis in mice to rats on gestation days (GD) 13 and 15. Blood and tissue data were analyzed using the extrapolated PBPK model that was modified to simulate 2-ME and 2-MAA kinetics in maternal plasma and total embryo tissues in pregnant rats. The original mouse model was simplified by combining the embryos and placenta with the richly perfused tissue compartment. The model includes a description of the growth of the developing embryo and changes in the physiology of the dam during pregnancy. Biotransformation pathways of 2-ME to either ethylene glycol (EG) or to 2-MAA were described as first-order processes based on the data collected from rats by Green et al., (Occup. Hyg. 2, 67-75, 1996). Tissue partition coefficients (PCs) for 2-ME and 2-MAA were determined for a variety of maternal tissues and the embryos. Model simulations closely reflected the biological measurement of 2-ME and 2-MAA concentrations in blood and embryo tissue following gavage or iv administration of 2-ME or 2-MAA. The PBPK model for rats as described here is well suited for extrapolation to pregnant women and for assessment of 2-MAA dosimetry under various conditions of possible human exposure to 2-ME.


Subject(s)
Acetates/pharmacokinetics , Ethylene Glycols/pharmacokinetics , Models, Biological , Pregnancy, Animal/metabolism , Teratogens/pharmacokinetics , Acetates/toxicity , Animals , Body Fluid Compartments/drug effects , Body Weight/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian/metabolism , Ethylene Glycols/toxicity , Female , Mammary Glands, Animal/drug effects , Mice , Organ Size/drug effects , Placenta/drug effects , Placenta/metabolism , Pregnancy , Rats , Teratogens/toxicity , Tissue Distribution
19.
Arch Toxicol ; 72(10): 609-21, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9851676

ABSTRACT

Trans- and cis-1,2-dichloroethylene (DCE) isomers inhibit their own metabolism in vivo by inactivation of the metabolizing enzyme, presumably the cytochrome P450 isoform, CYP2E1. In this study, we examined cytochrome P450 isoform-specific inhibition by three chloroethylenes, cis-DCE, trans-DCE, and trichloroethylene (TCE), and evaluated several kinetic mechanisms of enzyme inhibition with physiological models of inhibition. Trans-DCE was more potent than cis-DCE, and both were much more effective than TCE in inhibiting CYP2E1. The kinetics of in vitro loss of p-nitrophenol hydroxylase (pNP-OH) activity (a marker of CYP2E1) in microsomal incubations and of the in vivo gas uptake results were most consistent with a mechanism in which inhibition of the metabolizing enzyme (CYP2E1) was presumed to be related to interaction of a reactive DCE metabolite with remaining substrate-bound, active CYP2E1. The kinetics of inhibition by TCE, a weak inhibitor in vitro, were very different from that of the dichloroethylenes. With TCE, parent compound concentrations influenced enzyme loss. Trans-DCE was a more potent inhibitor of CYP2E1 than cis-DCE based on both in vivo and in vitro studies. Quantitative differences in the inhibitory properties of the 1,2-DCE isomers may be due to the different stability of epoxides formed from bioactivation by CYP2E1. Epoxide intermediates of DCE metabolism, reacting by water addition, would yield dialdehyde, a potent cross-linking reagent.


Subject(s)
Cytochrome P-450 CYP2E1 Inhibitors , Dichloroethylenes/pharmacology , Trichloroethylene/pharmacology , Vinyl Chloride/pharmacology , Animals , Cytochrome P-450 CYP2E1/metabolism , In Vitro Techniques , Male , Models, Chemical , Molecular Structure , Rats , Rats, Inbred F344 , Time Factors
20.
J Toxicol Environ Health ; 50(5): 433-49, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9140463

ABSTRACT

Chemicals such as cis-platinum, formaldehyde, chromate, copper, and certain arsenic compounds have been shown to produce DNA-protein cross-links in human in vitro cell systems at high doses, such as those in the cytotoxic range. Thus far there have only been a limited number of other chemicals evaluated for their ability to produce cross-links. The purpose of the work described here was to evaluate whether select industrial chemicals can form DNA-protein cross-links in human cells in vitro. We evaluated acetaldehyde, acrolein, diepoxybutane, paraformaldehyde, 2-furaldehyde, propionaldehyde, chloroacetaldehyde, sodium arsenite, and a deodorant tablet [Mega Blue; hazardous component listed as tris(hydroxymethyl)nitromethane]. Short- and long-term cytotoxicity was evaluated and used to select appropriate doses for in vitro testing. DNA-protein cross-linking was evaluated at no fewer than three doses and two cell lysate washing temperatures (45 and 65 degrees C) in Epstein-Barr virus (EBV) human Burkitt's lymphoma cells. The two washing temperatures were used to assess the heat stability of the DNA-protein cross-link, 2-Furaldehyde, acetaldehyde, and propionaldehyde produced statistically significant increases in DNA-protein cross-links at washing temperatures of 45 degrees C, but not 65 degrees C, and at or above concentrations of 5, 17.5, and 75 mM, respectively. Acrolein, diepoxybutane, paraformaldehyde, and Mega Blue produced statistically significant increases in DNA-protein cross-links washed at 45 and 65 degrees C at or above concentrations of 0.15 mM, 12.5 mM, 0.003%, and 0.1%, respectively. Sodium arsenite and chloroacetaldehyde did not produce significantly increased DNA-protein cross-links at either temperature nor at any dose tested. Excluding paraformaldehyde and 2-furaldehyde treatments, significant increases in DNA-protein cross-links were observed only at doses that resulted in complete cell death within 4 d following dosing. This work demonstrates that DNA-protein cross-links can be formed in vitro following exposure to a variety of industrial compounds and that most cross-links are formed at cytotoxic concentrations.


Subject(s)
Cross-Linking Reagents/toxicity , DNA-Binding Proteins/metabolism , DNA/metabolism , Acetaldehyde/analogs & derivatives , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Acrolein/metabolism , Acrolein/toxicity , Aldehydes/metabolism , Aldehydes/toxicity , Arsenates/metabolism , Arsenates/toxicity , Carcinogens/metabolism , Carcinogens/toxicity , Cell Death/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Cross-Linking Reagents/metabolism , DNA/drug effects , DNA-Binding Proteins/drug effects , Epoxy Compounds/metabolism , Epoxy Compounds/toxicity , Formaldehyde/metabolism , Formaldehyde/toxicity , Furaldehyde/metabolism , Furaldehyde/toxicity , Humans , Lymphoma/genetics , Lymphoma/pathology , Methane/analogs & derivatives , Methane/metabolism , Methane/toxicity , Mutagens/metabolism , Mutagens/toxicity , Nitroparaffins/metabolism , Nitroparaffins/toxicity , Polymers/metabolism , Polymers/toxicity , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...