Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Toxics ; 11(2)2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36851061

ABSTRACT

An array physiologically-based pharmacokinetic (PBPK) model represents a streamlined method to simultaneously quantify dosimetry of multiple compounds. To predict internal dosimetry of jet fuel components simultaneously, an array PBPK model was coded to simulate inhalation exposures to one or more selected compounds: toluene, ethylbenzene, xylenes, n-nonane, n-decane, and naphthalene. The model structure accounts for metabolism of compounds in the lung and liver, as well as kinetics of each compound in multiple tissues, including the cochlea and brain regions associated with auditory signaling (brainstem and temporal lobe). The model can accommodate either diffusion-limited or flow-limited kinetics (or a combination), allowing the same structure to be utilized for compounds with different characteristics. The resulting model satisfactorily simulated blood concentration and tissue dosimetry data from multiple published single chemical rat studies. The model was then utilized to predict tissue kinetics for the jet fuel hearing loss study (JTEH A, 25:1-14). The model was also used to predict rat kinetic comparisons between hypothetical exposures to JP-8 or a Virent Synthesized Aromatic Kerosene (SAK):JP-8 50:50 blend at the occupational exposure limit (200 mg/m3). The array model has proven useful for comparing potential tissue burdens resulting from complex mixture exposures.

2.
Arch Toxicol ; 87(4): 661-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314320

ABSTRACT

Multiple oximes have been synthesized and evaluated for use as countermeasures against chemical warfare nerve agents. The current U.S. military and civilian oxime countermeasure, 2-[(hydroxyimino)methyl]-1-methylpyridin-1-ium chloride (2-PAM), is under consideration for replacement with a more effective acetylcholinesterase reactivator, 1,1'-methylenebis{4-hydroxyiminomethyl}pyridinium dimethanesulfonate (MMB-4). Kinetic data in the scientific literature for MMB-4 are limited; therefore, a physiologically based pharmacokinetic (PBPK) model was developed for a structurally related oxime, 1,1'-trimethylenebis{4-hydroximinomethyl}pyridinium dibromide. Based on a previous model structure for the organophosphate diisopropylfluorophosphate, the model includes key sites of acetylcholinesterase inhibition (brain and diaphragm), as well as fat, kidney, liver, rapidly perfused tissues and slowly perfused tissues. All tissue compartments are diffusion limited. Model parameters were collected from the literature, predicted using quantitative structure-property relationships or, when necessary, fit to available pharmacokinetic data from the literature. The model was parameterized using rat plasma, tissue and urine time course data from intramuscular administration, as well as human blood and urine data from intravenous and intramuscular administration; sensitivity analyses were performed. The PBPK model successfully simulates rat and human data sets and has been evaluated by predicting intravenous mouse and intramuscular human data not used in the development of the model. Monte Carlo analyses were performed to quantify human population kinetic variability in the human evaluation data set. The model identifies potential pharmacokinetic differences between rodents and humans, indicated by differences in model parameters between species. The PBPK model can be used to optimize the dosing regimen to improve oxime therapeutic efficacy in a human population.


Subject(s)
Cholinesterase Reactivators/pharmacokinetics , Oximes/pharmacokinetics , Adult , Animals , Cholinesterase Reactivators/administration & dosage , Computer Simulation , Female , Humans , Injections, Intramuscular , Injections, Intravenous , Male , Mice , Middle Aged , Models, Biological , Monte Carlo Method , Rats , Rats, Wistar , Species Specificity , Tissue Distribution , Young Adult
3.
Inhal Toxicol ; 20(4): 375-90, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18302046

ABSTRACT

Acetaldehyde is an important intermediate in the chemical synthesis and normal oxidative metabolism of several industrially important compounds, including ethanol, ethyl acetate, and vinyl acetate. Chronic inhalation of acetaldehyde leads to degeneration of the olfactory and respiratory epithelium in rats at concentrations > 50 ppm (90 day exposure) and respiratory and olfactory nasal tumors at concentrations > or = 750 ppm, the lowest concentration tested in the 2-yr chronic bioassay. Differences in the anatomy and biochemistry of the rodent and human nose, including polymorphisms in human high-affinity acetaldehyde dehydrogenase (ALDH2), are important considerations for interspecies extrapolations in the risk assessment of acetaldehyde. A physiologically based pharmacokinetic model of rat and human nasal tissues was constructed for acetaldehyde to support a dosimetry-based risk assessment for acetaldehyde (Dorman et al., 2008). The rodent model was developed using published metabolic constants and calibrated using upper-respiratory-tract acetaldehyde extraction data. The human nasal model incorporates previously published tissue volumes, blood flows, and acetaldehyde metabolic constants. ALDH2 polymorphisms were represented in the human model as reduced rates of acetaldehyde metabolism. Steady-state dorsal olfactory epithelial tissue acetaldehyde concentrations in the rat were predicted to be 409, 6287, and 12,634 microM at noncytotoxic (50 ppm), and cytotoxic/tumorigenic exposure concentrations (750 and 1500 ppm), respectively. The human equivalent concentration (HEC) of the rat no-observed-adverse-effect level (NOAEL) of 50 ppm, based on steady-state acetaldehyde concentrations from continual exposures, was 67 ppm. Respiratory and olfactory epithelial tissue acetaldehyde and H(+) (pH) concentrations were largely linear functions of exposure in both species. The impact of presumed ALDH2 polymorphisms on human olfactory tissue concentrations was negligible; the high-affinity, low-capacity ALDH2 does not contribute significantly to acetaldehyde metabolism in the nasal tissues. The human equivalent acetaldehyde concentration for homozygous low activity was 66 ppm, 1.5% lower than for the homozygous full activity phenotype. The rat and human acetaldehyde PBPK models developed here can also be used as a bridge between acetaldehyde dose-response and mode-of-action data as well as between similar databases for other acetaldehyde-producing nasal toxicants.


Subject(s)
Acetaldehyde/pharmacokinetics , Air Pollutants, Occupational/pharmacokinetics , Aldehyde Dehydrogenase/genetics , Mitochondrial Proteins/genetics , Models, Biological , Nasal Cavity/metabolism , Polymorphism, Genetic , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Computer Simulation , Dose-Response Relationship, Drug , Humans , Inhalation Exposure , Mitochondrial Proteins/metabolism , Nasal Cavity/drug effects , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Rats
4.
J Toxicol Environ Health A ; 70(18): 1493-504, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17710610

ABSTRACT

Homeostatic mechanisms controlling uptake, storage, and elimination of dietary manganese (Mn) afford protection against fluctuations in tissue manganese (Mn) levels. Homeostatic control of inhaled Mn is less well understood, but important in assessing likely risks of Mn inhalation. Two compartmental kinetic models were used to characterize the influence of Mn exposure level and route (oral, inhalation) on uptake, elimination, and transport of Mn. The models were fitted to or used to interpret data from five whole-body Mn elimination studies: one dietary Mn balance study, two biliary elimination studies, and one acute and one chronic. As dietary Mn concentrations increased from low sufficiency (1.5 ppm) to sufficiency (20 ppm), control of Mn uptake shifts from the intestine (principally) to more proportional control by both intestinal tissues and liver. Using a two-compartment distribution model, the increased elimination of 54Mn tracer doses in response to increases in dietary Mn (rats and mice) or inhaled Mn (rats) resulted from elevation in Mn elimination rate constants rather than changes in intercompartmental transfer rate constants between a central compartment and deep compartment. The pharmacokinetic (PK) analysis also indicated differential control of absorption in single gavage oral dose studies versus continuous high oral doses in the feed. The gavage study indicated increased elimination rate constants, and the chronic study showed reduced rate constants for absorption. These dose dependencies in uptake and elimination are necessary inputs for comprehensive PK models guiding human health risk assessments with Mn.


Subject(s)
Manganese/pharmacokinetics , Models, Biological , Animals , Bile/chemistry , Dose-Response Relationship, Drug , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Mice , Rats , Rats, Inbred Strains
5.
J Toxicol Environ Health A ; 70(18): 1505-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17710611

ABSTRACT

Current concerns regarding inhalation exposure to Mn, a component from oxidation of the gasoline antiknock agent MMT, have stimulated interest in developing kinetic tools for describing the inhalation and combined inhalation/oral route kinetics of Mn. Kinetic approaches were integrated kinetic for (1) bulk tissue Mn kinetics and (2) hepato-intestinal control of oral-route Mn uptake into a integrated model structure connecting systemic and oral Mn. Linkages were developed between the hepato-intestinal and systemic tissues in order to evaluate differences in hepatic processing of orally absorbed Mn and systemic Mn. The integrated, unified model described the uptake, net absorption, and elimination of ingested Mn and the elimination kinetics of i.v. administered (systemic) Mn by treating Mn arriving at the liver from systemic versus portal blood differently. Hepatic extraction of orally absorbed Mn in rats predicted through simulation of the oral uptake data was 19, 54, and 78% at dietary exposures of 1.5, 11.2, and 100 ppm, respectively. In contrast, hepatic extraction of systemic Mn predicted through simulation of elimination kinetics i.v. tracer Mn was much less, 0.004, 0.005, or 0.009% at dietary levels of 2, 10, and 100 ppm, respectively. These differences in hepatic processing of blood Mn derived from different dose routes need to be accounted for in more complete PK models for Mn that are intended to support human health risk assessments.


Subject(s)
Manganese/pharmacokinetics , Models, Biological , Administration, Inhalation , Administration, Oral , Animals , Intestinal Absorption , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Rats , Rats, Inbred Strains
6.
J Toxicol Environ Health A ; 70(18): 1515-26, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17710612

ABSTRACT

Manganese (Mn), an essential metal nutrient, produces neurotoxicity in workers exposed chronically to high concentrations of Mn-containing dusts. Our long-term goal was to develop a physiologically based pharmacokinetic (PBPK) model to support health risk assessments for Mn. A PK model that accounts for Mn-tracer kinetics and steady-state tissue Mn in rats on normal diets (about 45 ppm Mn) is described. The focus on normal dietary intakes avoids inclusion of dose-dependent processes that maintain Mn homeostasis at higher dose rates. Data used for model development were obtained from published literature. The model represents six tissues: brain, respiratory tract, liver, kidneys, bone, and muscle. Each of these has a shallow tissue pool in rapid equilibration with blood and a deep tissue store, connected to the shallow pool by transfer rate constants. Intraperitoneal (i.p.) tracer Mn is absorbed into systemic blood and equilibrated with the shallow and deep pools of tissue Mn. The model was calibrated to match steady-state tissue concentrations and radiotracer kinetics following an i.p. dose of 54Mn. Successful simulations showed uptake of 0.8% of dietary Mn, and estimated tissue partition coefficients and transfer rate constants in the tissues. Inhalation tracer 54Mn studies could only be adequately modeled by assuming that deposited Mn was absorbed into deep tissue stores in the lung before becoming available to move via blood to other tissues. In summary, this present effort provides the basic structure of a multiroute PBPK model for Mn that should now be easily extended to include homeostatic control and inhalation exposures in order to support risk assessment calculations for Mn.


Subject(s)
Manganese/pharmacokinetics , Models, Biological , Radioisotopes/pharmacokinetics , Administration, Inhalation , Administration, Oral , Animals , Bone and Bones/metabolism , Brain/metabolism , Kidney/metabolism , Kinetics , Liver/metabolism , Male , Radioactive Tracers , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Respiratory System/metabolism
7.
J Toxicol Environ Health A ; 70(5): 445-64, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17454569

ABSTRACT

The potential associations between exposure to nickel compounds and cancer have been evaluated in both animal and epidemiological studies of occupationally exposed workers. The results of the epidemiological studies suggest that not all nickel compounds are equally carcinogenic, an observation supported by the animal bioassay results. Given the complexity and the differences in the modes of uptake of different forms of nickel by cells and the subsequent delivery of nickel to the nucleus, it would be expected that some forms of nickel would be more potent than others. A physiologically based pharmacokinetic (PBPK) model would be useful in estimating the cellular exposure to nickel resulting from inhalation of the different forms of nickel. To this end, a preliminary model of a tracheobronchial epithelial cell was developed to describe the differences in the extracellular and intracellular kinetics of the different classes of nickel compounds. Data available in the published literature were used to define the initial model parameters. The resulting cellular dosimetry model was able to describe kinetic data on three forms of nickel (soluble chloride and insoluble sulfide and subsulfide). This preliminary model development effort has identified critical data gaps that could be filled by additional research. The ultimate goal will be to integrate a refined cellular dosimetry model with published lung deposition/clearance and systemic distribution/clearance models for nickel. The use of such an integrated PBPK model would allow for more biologically based risk estimates for the inhalation of the different nickel compounds, as well as mixtures of these compounds.


Subject(s)
Models, Biological , Nickel/pharmacokinetics , Respiratory Mucosa/metabolism , Aerosols , Animals , Dose-Response Relationship, Drug , Humans , Inhalation Exposure , Phagocytosis/drug effects , Respiratory Mucosa/cytology , Risk Assessment
8.
Regul Toxicol Pharmacol ; 47(1): 1-18, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16901594

ABSTRACT

The current Public Health Goal (PHG) for perchloroethylene (PCE) was derived using upper-bound estimates of fractional PCE metabolism in humans. These estimates were in part obtained from a published evaluation of the uncertainty and variability in human PCE metabolism conducted using a physiologically-based pharmacokinetic (PBPK) model in a Markov chain Monte Carlo (MCMC) analysis; however, the data used in that analysis were limited to post-exposure PCE blood and exhaled air concentrations from a single study. A more recent study [Volkel, W., Friedewald, M., Lederer, E., Pahler, A., Parker, J., Dekant, W., 1998. Biotransformation of perchloroethene: dose-dependent excretion of trichloroacetic acid, dichloroacetic acid, and N-acetyl-S-(trichlorovinyl)-l-cysteine in rats and humans after inhalation. Toxicol. Appl. Pharmacol. 153(1), 20-27.] provides data on blood concentrations of PCE and its major metabolite, trichloroacetic acid (TCA), and urinary excretion of TCA following exposure of human subjects to lower concentrations of PCE (10-40ppm) than in previous studies. In the present effort, a new MCMC analysis was performed that focused on data from this study along with two others [Fernandez, J., Guberan, E., Caperos, J., 1976. Experimental human exposures to tetrachloroethylene vapor and elimination in breath after inhalation. Am. Ind. Hyg. Assoc. J. 37, 143-150; Monster, A., Boersma, G., Steenweg, H., 1979. Kinetics of tetrachloroethylene in volunteers; influence of exposure concentration and work load. Int. Arch. Occup. Environ. Health 42, 303-309.] providing data on PCE blood concentrations and urinary excretion of TCA. To provide an accurate prediction of TCA kinetics, the PBPK model used here includes a description of the metabolism of PCE to TCA in both the liver and kidney. The resulting upper 95th percentile estimates of fraction of PCE metabolized by inhalation and oral routes were 2.1 and 5.2%, respectively, compared to 58 and 79% used in the derivation of the PHG.


Subject(s)
Carcinogens, Environmental/adverse effects , Markov Chains , Monte Carlo Method , Public Health , Tetrachloroethylene/adverse effects , Carcinogens, Environmental/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Inhalation Exposure , Male , Models, Biological , Models, Statistical , Risk Assessment/statistics & numerical data , Tetrachloroethylene/pharmacokinetics , Uncertainty
9.
Regul Toxicol Pharmacol ; 45(1): 55-65, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16439044

ABSTRACT

An updated PBPK model of methylene chloride (DCM, dichloromethane) carcinogenicity in mice was recently published using Bayesian statistical methods (Marino et al., 2006). In this work, this model was applied to humans, as recommended by Sweeney et al.(2004). Physiological parameters for input into the MCMC analysis were selected from multiple sources reflecting, in each case, the source that was considered to represent the most current scientific evidence for each parameter. Metabolic data for individual subjects from five human studies were combined into a single data set and population values derived using MCSim. These population values were used for calibration of the human model. The PBPK model using the calibrated metabolic parameters was used to perform a cancer risk assessment for DCM, using the same tumor incidence and exposure concentration data relied upon in the current IRIS entry. Unit risks, i.e., the risk of cancer from exposure to 1 microg/m3 over a lifetime, for DCM were estimated using the calibrated human model. The results indicate skewed distributions for liver and lung tumor risks, alone or in combination, with a mean unit risk (per microg/m3) of 1.05 x 10(-9), considering both liver and lung tumors. Adding the distribution of genetic polymorphisms for metabolism to the ultimate carcinogen, the unit risks range from 0 (which is expected given that approximately 20% of the US population is estimated to be nonconjugators) up to a unit risk of 2.70 x 10(-9) at the 95th percentile. The median, or 50th percentile, is 9.33 x 10(-10), which is approximately a factor of 500 lower than the current EPA unit risk of 4.7 x 10(-7) using a previous PBPK model. These values represent the best estimates to date for DCM cancer risk because all available human data sets were used, and a probabilistic methodology was followed.


Subject(s)
Carcinogens/pharmacokinetics , Methylene Chloride/pharmacokinetics , Models, Biological , Neoplasms/chemically induced , Carcinogens/toxicity , Dose-Response Relationship, Drug , Glutathione Transferase/genetics , Humans , Inhalation Exposure , Markov Chains , Methylene Chloride/toxicity , Monte Carlo Method , Neoplasms/genetics , Polymorphism, Genetic , Risk Assessment
10.
Regul Toxicol Pharmacol ; 45(1): 44-54, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16442684

ABSTRACT

The current USEPA cancer risk assessment for dichloromethane (DCM) is based on deterministic physiologically based pharmacokinetic (PBPK) modeling involving comparative metabolism of DCM by the GST pathway in the lung and liver of humans and mice. Recent advances in PBPK modeling include probabilistic methods and, in particular, Bayesian inference to quantitatively address variability and uncertainty separately. Although Bayesian analysis of human PBPK models has been published, no such efforts have been reported specifically addressing the mouse, apart from results included in the OSHA final rule on DCM. Certain aspects of the OSHA model, however, are not consistent with current approaches or with the USEPA's current DCM cancer risk assessment. Therefore, Bayesian analysis of the mouse PBPK model and dose-response modeling was undertaken to support development of an improved cancer risk assessment for DCM. A hierarchical population model was developed and prior parameter distributions were selected to reflect parameter values that were considered the most appropriate and best available. Bayesian modeling was conducted using MCSim, a publicly available software program for Markov Chain Monte Carlo analysis. Mean posterior values from the calibrated model were used to develop internal dose metrics, i.e., mg DCM metabolized by the GST pathway/L tissue/day in the lung and liver using exposure concentrations and results from the NTP mouse bioassay, consistent with the approach used by the USEPA for its current DCM cancer risk assessment. Internal dose metrics were 3- to 4-fold higher than those that support the current USEPA IRIS assessment. A decrease of similar magnitude was also noted in dose-response modeling results. These results show that the Bayesian PBPK model in the mouse provides an improved basis for a cancer risk assessment of DCM.


Subject(s)
Carcinogens/pharmacokinetics , Methylene Chloride/pharmacokinetics , Models, Biological , Neoplasms/chemically induced , Animals , Bayes Theorem , Dose-Response Relationship, Drug , Inhalation Exposure , Markov Chains , Mice , Monte Carlo Method , Risk Assessment
11.
J Toxicol Environ Health A ; 68(5): 329-51, 2005 Mar 12.
Article in English | MEDLINE | ID: mdl-15799626

ABSTRACT

Several chronic bioassays have been conducted in multiple strains of mice in which various concentrations of arsenate or arsenite were administered in the drinking water without a tumorigenic effect. However, one study (Ng et al., 1999) reported a significant increase in tumor incidence in C57Bl/6J mice exposed to arsenic in their drinking water throughout their lifetime, with no tumors reported in controls. A physiologically based pharmacokinetic model for arsenic in the mouse has previously been developed (Gentry et al., 2004) to investigate potential differences in tissue dosimetry of arsenic species across various strains of mice. Initial results indicated no significant differences in blood, liver, or urine dosimetry in B6C3F1 and C57Bl/6 mice for acute or subchronic exposure. The current work was conducted to compare model-predicted estimates of tissue dosimetry to additional kinetic information from the (C57Bl/6 xCBA)F1 and TgAc mouse. The results from the current modeling indicate that the pharmacokinetic parameters derived based on information in the B6C3F1 mouse adequately describe the measured concentrations in the blood/plasma, liver, and urine of both the (C57Bl/6 x CBA)F1 and TgAc mouse, providing further support that the differences in response observed in the chronic bioassays are not related to strain-specific differences in pharmacokinetics. One significant finding was that no increases in skin or lung concentrations of arsenic species in the (C57Bl/6 x CBA)F1 strain were observed following administration of low concentrations (0.2 or 2 mg/U of arsenate in the drinking water, even though differences in response in the skin were reported. These data suggest that pharmacodynamic changes may be observed following exposure to arsenic compounds without an observable change in tissue dosimetry. These results provided further indirect support for the existence of inducible arsenic efflux in these tissues.


Subject(s)
Arsenic/pharmacokinetics , Arsenic/toxicity , Models, Theoretical , Administration, Oral , Animals , Arsenic/administration & dosage , Biological Assay , Drug Administration Schedule , Forecasting , Mice , Mice, Inbred C57BL , Reproducibility of Results , Skin Neoplasms/chemically induced , Skin Neoplasms/veterinary , Tissue Distribution , Water Supply
12.
Toxicol Sci ; 85(2): 823-38, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15746009

ABSTRACT

Bisphenol A (BPA) is a weakly estrogenic monomer used in the production of polycarbonate plastic and epoxy resins, both of which are used in food contact and other applications. A physiologically based pharmacokinetic (PBPK) model of BPA pharmacokinetics in rats and humans was developed to provide a physiological context in which the processes controlling BPA pharmacokinetics (e.g., plasma protein binding, enterohepatic recirculation of the glucuronide [BPAG]) could be incorporated. A uterine tissue compartment was included to allow the correlation of simulated estrogen receptor (ER) binding of BPA with increases in uterine wet weight (UWW) in rats. Intravenous- and oral-route blood kinetics of BPA in rats and oral-route plasma and urinary elimination kinetics in humans were well described by the model. Simulations of rat oral-route BPAG pharmacokinetics were less exact, most likely the result of oversimplification of the GI tract compartment. Comparison of metabolic clearance rates derived from fitting rat i.v. and oral-route data implied that intestinal glucuronidation of BPA is significant. In rats, but not humans, terminal elimination rates were strongly influenced by enterohepatic recirculation. In the absence of BPA binding to plasma proteins, simulations showed high ER occupancy at doses without uterine effects. Restricting free BPA to the measured unbound amount demonstrated the importance of including plasma binding in BPA kinetic models: the modeled relationship between ER occupancy and UWW increases was consistent with expectations for a receptor-mediated response with low ER occupancy at doses with no response and increasing occupancy with larger increases in UWW.


Subject(s)
Estrogens, Non-Steroidal/administration & dosage , Estrogens, Non-Steroidal/pharmacokinetics , Phenols/administration & dosage , Phenols/pharmacokinetics , Uterus/metabolism , Administration, Oral , Algorithms , Animals , Benzhydryl Compounds , Blood Proteins/metabolism , Dose-Response Relationship, Drug , Enterohepatic Circulation , Female , Glucuronides/metabolism , Humans , Injections, Intravenous , Male , Models, Statistical , Organ Size/drug effects , Protein Binding , Rats , Rats, Inbred F344 , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism
13.
Toxicol Sci ; 79(2): 381-93, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15056818

ABSTRACT

The physiological and biochemical processes that determine the tissue concentration time courses (pharmacokinetics) of xenobiotics vary, in some cases significantly, with age and gender. While it is known that age- and gender-specific differences have the potential to affect tissue concentrations and, hence, individual risk, the relative importance of the contributing processes and the quantitative impact of these differences for various life stages are not well characterized. The objective of this study was to identify age- and gender-specific differences in physiological and biochemical processes that affect tissue dosimetry and integrate them into a predictive physiologically based pharmacokinetic (PBPK) life-stage model. The life-stage model was exercised for several environmental chemicals with a variety of physicochemical, biochemical, and mode-of-action properties. In general, predictions of average pharmacokinetic dose metrics for a chemical across life stages were within a factor of two, although larger transient variations were predicted, particularly during the neonatal period. The most important age-dependent pharmacokinetic factor appears to be the potential for decreased clearance of a toxic chemical in the perinatal period due to the immaturity of many metabolic enzyme systems, although this same factor may also reduce the production of a reactive metabolite. Given the potential for age-dependent pharmacodynamic factors during early life, there may be chemicals and health outcomes for which decreased clearance over a relatively brief period could have a substantial impact on risk.


Subject(s)
Models, Biological , Xenobiotics/pharmacokinetics , 2-Propanol/pharmacokinetics , Adolescent , Adult , Age Factors , Aged , Biotransformation , Body Burden , Child , Female , Humans , Infant , Infant, Newborn , Male , Methylene Chloride/pharmacokinetics , Nicotine/pharmacokinetics , Polychlorinated Dibenzodioxins/pharmacokinetics , Sex Factors , Tetrachloroethylene/pharmacokinetics , Tissue Distribution , Vinyl Chloride/pharmacokinetics
14.
J Toxicol Environ Health A ; 67(1): 43-71, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14668111

ABSTRACT

A remarkable feature of the carcinogenicity of inorganic arsenic is that while human exposures to high concentrations of inorganic arsenic in drinking water are associated with increases in skin, lung, and bladder cancer, inorganic arsenic has not typically caused tumors in standard laboratory animal test protocols. Inorganic arsenic administered for periods of up to 2 yr to various strains of laboratory mice, including the Swiss CD-1, Swiss CR:NIH(S), C57Bl/6p53(+/-), and C57Bl/6p53(+/+), has not resulted in significant increases in tumor incidence. However, Ng et al. (1999) have reported a 40% tumor incidence in C57Bl/6J mice exposed to arsenic in their drinking water throughout their lifetime, with no tumors reported in controls. In order to investigate the potential role of tissue dosimetry in differential susceptibility to arsenic carcinogenicity, a physiologically based pharmacokinetic (PBPK) model for inorganic arsenic in the rat, hamster, monkey, and human (Mann et al., 1996a, 1996b) was extended to describe the kinetics in the mouse. The PBPK model was parameterized in the mouse using published data from acute exposures of B6C3F1 mice to arsenate, arsenite, monomethylarsonic acid (MMA), and dimethylarsinic acid (DMA) and validated using data from acute exposures of C57Black mice. Predictions of the acute model were then compared with data from chronic exposures. There was no evidence of changes in the apparent volume of distribution or in the tissue-plasma concentration ratios between acute and chronic exposure that might support the possibility of inducible arsenite efflux. The PBPK model was also used to project tissue dosimetry in the C57Bl/6J study, in comparison with tissue levels in studies having shorter duration but higher arsenic treatment concentrations. The model evaluation indicates that pharmacokinetic factors do not provide an explanation for the difference in outcomes across the various mouse bioassays. Other possible explanations may relate to strain-specific differences, or to the different durations of dosing in each of the mouse studies, given the evidence that inorganic arsenic is likely to be active in the later stages of the carcinogenic process.


Subject(s)
Arsenic Poisoning/complications , Arsenic , Carcinogens , Disease Models, Animal , Models, Chemical , Neoplasms/chemically induced , Water Pollutants, Chemical , Acute Disease , Administration, Oral , Animals , Arsenates/pharmacokinetics , Arsenates/toxicity , Arsenic/pharmacokinetics , Arsenic/toxicity , Arsenic Poisoning/metabolism , Arsenicals/adverse effects , Arsenicals/pharmacokinetics , Arsenites/pharmacokinetics , Arsenites/toxicity , Cacodylic Acid/pharmacokinetics , Cacodylic Acid/toxicity , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Chronic Disease , Dose-Response Relationship, Drug , Environmental Exposure/adverse effects , Incidence , Mice , Mice, Inbred C57BL , Neoplasms/epidemiology , Predictive Value of Tests , Tissue Distribution , Water Pollutants, Chemical/pharmacokinetics , Water Pollutants, Chemical/toxicity
15.
J Toxicol Environ Health A ; 66(23): 2209-25, 2003 Dec 12.
Article in English | MEDLINE | ID: mdl-14612334

ABSTRACT

Recent health risk assessments to propose a Reference Dose (RfD) for acetone (Forsyth, 2001; U.S. EPA, 2001) have been based on the results of an oral subchronic study conducted in rats and mice (Dietz et al., 1991; NTP, 1991). These assessments have utilized the traditional concept of establishing the RfD by determining the lowest experimentally determined No-Observed-Adverse-Effect Level (NOAEL) and applying various Uncertainty Factors (UFs) (U.S. EPA, 1988). This article describes a risk assessment for acetone based on the systemic toxicity observed in subchronic and developmental toxicity studies to estimate an RfD and an inhalation reference concentration (RfC) for acetone. Specifically, this approach examined the subchronic study by Dietz et al. (1991), as well as an inhalation developmental toxicity study on acetone (Mast et al., 1988) and several toxicology studies of isopropanol (IPA). This was accomplished by applying a physiologically based pharmacokinetic (PBPK) model developed previously for IPA and its metabolite acetone (Clewell et al., 2001). The incorporation of the PBPK model into the derivation of an RfD and RfC for acetone allowed for a tissue-based approach rather than an external exposure-based approach, making it possible to derive an oral RfD from an inhalation study. In addition, the use of the PBPK model to analyze data from chronic and reproductive/developmental studies conducted with IPA enabled an assessment of the potential for acetone to produce any of the effects observed in the IPA studies. This analysis provided sufficient information to reduce the need for UFs in the adjustment of the NOAEL from the oral subchronic study for the determination of an RfD. Using the PBPK model in the acetone risk assessment supports a composite UF of 60 for the subchronic study, compared to composite factors of 300 to 3000 in the other recent risk assessments. This difference resulted in an RfD of 16 mg/kg/d, compared to the values of 0.3 to 3 that have previously been estimated (Forsyth, 2001; U.S. EPA, 2001). Considering the results from the inhalation developmental study (Mast et al., 1988) resulted in an RfD of 8.7 mg/kg/d. Using this study also fills a data gap for acetone that exists if only the oral database for acetone is considered for RfD derivation. An RfC of 29 ppm was also estimated for acetone using the Mast et al. (1988) study results in combination with the PBPK model. The potential impact of endogenous acetone on a risk assessment for acetone is also discussed.


Subject(s)
Acetone/pharmacokinetics , Acetone/toxicity , Inhalation Exposure , Models, Theoretical , Solvents/pharmacokinetics , Solvents/toxicity , Administration, Oral , Animals , Humans , No-Observed-Adverse-Effect Level , Rats , Reference Values , Risk Assessment
16.
Inhal Toxicol ; 15(10): 987-1016, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12928976

ABSTRACT

In recent years, there have been growing concerns that due to differences, both pharmacokinetic and pharmacodynamic, between children and adults, children could be at greater risk of adverse effects following chemical exposure. The specific goal of this study was to demonstrate an approach for using physiologically based pharmacokinetic (PBPK) modeling to compare inhalation dosimetry in the adult and the child of both males and females. Three categories of gases were considered: rapidly and irreversibly reactive in the respiratory tract (ozone), relatively water-soluble and nonreactive (isopropanol), and relatively water-insoluble and nonreactive (styrene, vinyl chloride, and perchloroethylene). The nonreactive chemicals were also selected because they are metabolized in the respiratory tract. The age-related changes observed for the estimated dose metrics were a function of the physiochemical properties of the inhaled vapor and their interactions in the body. Blood concentrations estimated for all vapors, either poorly metabolized (e.g., PERC), moderately metabolized (e.g., ST), or highly metabolized vapors (e.g., IPA and VC), varied less than a factor of two between infants and adults. These changes, moreover, were confined to the first year after birth, a relatively short window compared to the total lifespan of the individual. In contrast, circulating metabolite concentrations estimated in the blood, as well as amounts metabolized in the liver and lung, appeared to be a strong function of age, due to their dependence on the maturity of the pertinent metabolic enzyme systems.


Subject(s)
2-Propanol/pharmacokinetics , Inhalation Exposure , Lung/anatomy & histology , Models, Theoretical , Oxidants, Photochemical/pharmacokinetics , Ozone/pharmacokinetics , Solvents/pharmacokinetics , Styrene/pharmacokinetics , Tetrachloroethylene/pharmacokinetics , Vinyl Chloride/pharmacokinetics , Adolescent , Adult , Age Factors , Aged , Child , Child, Preschool , Female , Gases , Humans , Infant , Infant, Newborn , Male , Middle Aged , Sex Factors , Volatilization
17.
Regul Toxicol Pharmacol ; 38(1): 1-16, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12878049

ABSTRACT

In recent years efforts have increased to develop a framework for assessing differences, both pharmacokinetic and pharmacodynamic, between children and adults for purposes of assessing risk of adverse effects following chemical exposure. The specific goal of this study was to demonstrate an approach for using PBPK modeling to compare maternal and fetal/neonatal blood and tissue dose metrics during pregnancy and lactation. Six chemical classes were targeted to provide a variety of physicochemical properties (volatility, lipophilicity, water solubility), and surrogate chemicals were selected to represent each class (isopropanol, vinyl chloride, methylene chloride, perchloroethylene, nicotine, and TCDD), based on the availability of pharmacokinetic information. These chemicals were also selected to provide different pharmacokinetic characteristics, including metabolic production of stable or reactive intermediates in the liver and competing pathways for metabolism. Changes in dosimetry during pregnancy predicted by the modeling were mainly attributable to the development of enzymatic pathways in the fetus or to changes in tissue composition in the mother and fetus during pregnancy. In general, blood concentrations were lower in the neonate during the lactation period than in the fetus during gestation. This postnatal decrease varied from only a slight change (for TCDD) to approximately four orders of magnitude (for vinyl chloride). As compared to maternal exposure, fetal/neonatal exposures ranged from approximately twice as great (for TCDD) to several orders of magnitude lower (for isopropanol). The results of this study are in general agreement with the analyses of data on pharmaceutical chemicals, which have suggested that the largest difference in pharmacokinetics observed between children and adults is for the perinatal period. The most important factor appears to be the potential for decreased clearance of toxic chemicals in the perinatal period due to immature metabolic enzyme systems, although this same factor can also reduce the risk from reactive metabolites during the same period.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Environmental Pollutants/adverse effects , Lactation/metabolism , Maternal-Fetal Exchange , Pharmacokinetics , Pregnancy Complications , Animals , Breast Feeding , Carcinogens/pharmacokinetics , Female , Fetus/drug effects , Humans , Infant, Newborn , Models, Biological , Pregnancy , Tissue Distribution , Toxicology/methods
18.
Regul Toxicol Pharmacol ; 36(1): 51-68, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12383718

ABSTRACT

An interspecies physiologically based pharmacokinetic (PBPK) model describing isopropanol (IPA) and its major metabolite, acetone, was applied to perform route-to-route and cross-species dosimetry to derive reference dose (RfD) and reference concentration (RfC) values for IPA. Adult PBPK models for rats and humans were extended to simulate exposure to IPA during pregnancy and used to estimate internal dose metrics in the mother and fetus during development. Endpoints from chronic, developmental, and reproductive toxicity studies were considered for the derivation of RfDs and RfCs. Due to uncertainties in the mode of action of toxicity for IPA and acetone, the dose metric used for most responses was the total area under the blood concentration curve (AUC) for the combination of IPA and acetone. This combined dose metric provided a more conservative estimate than those based on AUCs for IPA or acetone. Peak blood concentration of IPA was the dose metric for neurobehavioral effects. The recommended RfD and RfC for IPA are 10 mg/kg/day and 40 ppm, respectively, based on decreased fetal body weights. All of the PBPK-derived RfD or RfC values for various endpoints were similar (within a factor of 3), regardless of route of exposure in the animal study.


Subject(s)
2-Propanol/pharmacokinetics , 2-Propanol/toxicity , Models, Biological , Reproduction/drug effects , 2-Propanol/administration & dosage , Acetone/administration & dosage , Acetone/pharmacokinetics , Acetone/toxicity , Adult , Algorithms , Animals , Area Under Curve , Decision Trees , Dose-Response Relationship, Drug , Drug Administration Routes , Embryonic and Fetal Development/drug effects , Female , Fetus/drug effects , Humans , No-Observed-Adverse-Effect Level , Pregnancy , Rats , Reference Values , Reproduction/physiology , Risk Assessment/methods , Toxicity Tests/standards , Toxicity Tests, Chronic , Uncertainty
SELECTION OF CITATIONS
SEARCH DETAIL
...