Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 132: 105185, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35537634

RESUMEN

The Steering Committee of the Alliance for Risk Assessment (ARA) opened a call for scientists interested in resolving what appeared to be a conundrum in estimating of the half-life of perfluorooctanoate (PFOA) in humans. An Advisory Committee was formed from nominations received and a subsequent invitation led to the development of three small independent working groups to review appropriate information and attempt a resolution. Initial findings were shared among these groups and a conclusion developed from the ensuing discussions. Many human observational studies have estimated the PFOA half-life. Most of these studies note the likely occurrence of unmonitored PFOA exposures, which could inflate values of the estimated PFOA half-life. Also, few of these studies estimated the half-life of PFOA isomers, the branched chains of which likely have shorter half-lives. This could deflate values of the estimated linear PFOA half-life. Fortunately, several studies informed both of these potential problems. The majority opinion of this international collaboration is that the studies striking the best balance in addressing some of these uncertainties indicate the likely central tendency of the human PFOA half-life is less than 2 years. The single best value appears to be the geometric mean (GM) of 1.3 years (Zhang et al., 2013, Table 3), based on a GM = 1.7 years in young females (n = 20) and GM = 1.2 years in males of all ages and older females (n = 66). However, a combined median value from Zhang et al. (2013) of 1.8 years also adds value to this range of central tendency. While the Collaboration found this study to be the least encumbered with unmonitored PFOA exposures and branched isomers, more studies of similar design would be valuable. Also valuable would be clarification around background exposures in other existing studies in case adjustments to half-life estimates are attempted.


Asunto(s)
Caprilatos , Fluorocarburos , Caprilatos/toxicidad , Femenino , Fluorocarburos/toxicidad , Semivida , Humanos , Masculino , Medición de Riesgo
2.
Toxicol Appl Pharmacol ; 417: 115463, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33631232

RESUMEN

By extending our Paraquat (PQ) work to include primates we have implemented a modelling and simulation strategy that has enabled PQ pharmacokinetic data to be integrated into a single physiologically based pharmacokinetic (PBPK) model that enables more confident extrapolation to humans. Because available data suggested there might be differences in PQ kinetics between primates and non-primates, a radiolabelled study was conducted to characterize pharmacokinetics and excretion in Cynomolgus monkeys. Following single intravenous doses of 0.01 or 0.1 mg paraquat dichloride/kg bw, plasma PQ concentration-time profiles were dose-proportional. Excretion up to 48 h (predominantly urinary) was 82.9%, with ca. 10% remaining unexcreted. In vitro blood binding was similar across Cynomolgus monkeys, humans and rat. Our PBPK model for the rat, mouse and dog, employing a single set of PQ-specific parameters, was scaled to Cynomolgus monkeys and well represented the measured plasma concentration-time profiles over 14 days. Addition of a cartilage compartment to the model better captured the percent remaining in the monkeys at 48 h, whilst having negligible effect on model predictions for the other species. The PBPK model performed well for all four species, demonstrating there is little difference in PQ kinetics between non-primates and primates enabling a more confident extrapolation to humans. Scaling of the PBPK model to humans, with addition of a human-specific dermal submodel based on in vitro human dermal absorption data, provides a valuable tool that could be employed in defining internal dosimetry to complement human health risk assessments.


Asunto(s)
Herbicidas/farmacocinética , Modelos Biológicos , Paraquat/farmacocinética , Animales , Simulación por Computador , Herbicidas/administración & dosificación , Herbicidas/sangre , Herbicidas/toxicidad , Humanos , Infusiones Intravenosas , Eliminación Intestinal , Macaca fascicularis , Paraquat/administración & dosificación , Paraquat/sangre , Paraquat/toxicidad , Ratas , Eliminación Renal , Medición de Riesgo , Absorción Cutánea , Especificidad de la Especie , Distribución Tisular , Toxicocinética
3.
Toxicol Appl Pharmacol ; 417: 115462, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33631233

RESUMEN

Paraquat dichloride (PQ) is a non-selective herbicide which has been the subject of numerous toxicology studies over more than 50 years. This paper describes the development of a physiologically-based pharmacokinetic (PBPK) model of PQ kinetics for the rat, mouse and dog, firstly to aid the interpretation of studies in which no kinetic measurements were made, and secondly to enable the future extension of the model to humans. Existing pharmacokinetic data were used to develop a model for the rat and mouse. Simulations with this preliminary model were then used to identify key data gaps and to design a new blood binding study to reduce uncertainty in critical aspects of the model. The new data provided evidence to support the model structure, and its predictive performance was then assessed against dog and rat datasets not used in model development. The PQ-specific model parameters are the same for all three species, with only the physiological parameters varying between species. This consistency across species provides a strong basis for extrapolation to other species, as demonstrated here for the dog. The model enables a wide range of PQ data to be linked together to provide a broad understanding of PQ pharmacokinetics in rodents and the dog, showing that the key aspects of PQ kinetics in these species are understood and adequately encapsulated within the model.


Asunto(s)
Herbicidas/farmacocinética , Modelos Biológicos , Paraquat/farmacocinética , Animales , Simulación por Computador , Perros , Herbicidas/sangre , Herbicidas/toxicidad , Eliminación Intestinal , Ratones , Paraquat/sangre , Paraquat/toxicidad , Unión Proteica , Ratas , Eliminación Renal , Medición de Riesgo , Especificidad de la Especie , Distribución Tisular , Toxicocinética
4.
Int J Environ Health Res ; 31(8): 951-962, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31850798

RESUMEN

A physiologically based pharmacokinetic (PBPK) model was developed to described uptake, disposition and clearance of bromate in the rat using published experimental data in rat. The rodent bromate model was extrapolated to human using species-specific physiological parameters and standard interspecies scaling of rate constants. The bromate model is kinetically linear (i.e. AUC and Cmax) across the range of drinking water concentrations used in the cancer bioassays (15 to 500 ppm). This is likely the result of the poor oral bioavailability of bromate due to high reduction rates in the intestinal tract. The bromate PBPK model was used to assess the human equivalent drinking water concentration (HEC) consistent with average plasma concentrations in the rodent bioassays. At drinking water concentrations <500 mg/L, the predicted HEC was two to three fold lower than the bioassay concentration and was dependent on the reported drinking water intake reported in the bioassay.


Asunto(s)
Bromatos/farmacocinética , Agua Potable/química , Contaminantes Químicos del Agua/farmacocinética , Animales , Disponibilidad Biológica , Bromatos/análisis , Simulación por Computador , Exposición Dietética/análisis , Femenino , Humanos , Modelos Biológicos , Ratas , Contaminantes Químicos del Agua/análisis
5.
Environ Res ; 182: 109017, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31865168

RESUMEN

Exposure to perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) has been associated with the occurrence of thyroid disease in some epidemiologic studies. We hypothesized that in a specific epidemiologic study based on the National Health and Nutrition Examination Survey, the association of subclinical thyroid disease with serum concentration of PFOA and PFOS was due to reverse causality. Thyroid hormone affects glomerular filtration, which in turn affects excretion of PFOA and PFOS. We evaluated this by linking a model of thyroid disease status over the lifetime to physiologically based pharmacokinetic models of PFOA and PFOS. Using Monte Carlo methods, we simulated the target study population and analyzed the data using multivariable logistic regression. The target and simulated populations were similar with respect to age, estimated glomerular filtration rate, serum concentrations of PFOA and PFOS, and prevalence of subclinical thyroid disease. Our findings suggest that in the target study the associations with subclinical hypothyroidism were overstated and the results for subclinical hyperthyroidism were, in general, understated. For example, for subclinical hypothyroidism in men, the reported odds ratio per ln(PFOS) increase was 1.98 (95% CI 1.19-3.28), whereas in the simulated data the bias due to reverse causality gave an odds ratio of 1.19 (1.16-1.23). Our results provide evidence of bias due to reverse causality in a specific cross-sectional study of subclinical thyroid disease with exposure to PFOA and PFOS among adults.


Asunto(s)
Ácidos Alcanesulfónicos , Contaminantes Ambientales , Fluorocarburos , Enfermedades de la Tiroides , Adulto , Caprilatos , Estudios Transversales , Contaminantes Ambientales/sangre , Contaminantes Ambientales/toxicidad , Femenino , Fluorocarburos/sangre , Fluorocarburos/toxicidad , Humanos , Masculino , Encuestas Nutricionales , Enfermedades de la Tiroides/inducido químicamente
6.
J Toxicol Environ Health A ; 83(19-20): 631-648, 2020 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-32757748

RESUMEN

A physiologically based pharmacokinetic (PBPK) model for di-isononyl phthalate (DiNP) was developed by adapting the existing models for di(2-ethylhexyl) phthalate (DEHP) and di-butylphthalate (DBP). Both pregnant rat and human time-course plasma and urine data were used to address the hydrolysis of DiNP in intestinal tract, plasma, and liver as well as hepatic oxidative metabolism and conjugation of the monoester and primary oxidative metabolites. Data in both rats and humans were available to inform the uptake and disposition of mono-isononyl phthalate (MiNP) as well as the three primary oxidative metabolites including hydroxy (7-OH)-, oxo (7-OXO)-, and carboxy (7-COX)-monoisononyl phthalate in plasma and urine. The DiNP model was reliable over a wide range of exposure levels in the pregnant rat as well as the two low exposure levels in humans including capturing the nonlinear behavior in the pregnant rat after repeated 750 mg/kg/day dosing. The presented DiNP PBPK model in pregnant rat and human, based upon an extensive kinetic dataset in both species, may provide a basis for assessing human equivalent exposures based upon either rodent or in vitro points of departure.


Asunto(s)
Contaminantes Ambientales/farmacocinética , Ácidos Ftálicos/farmacocinética , Plastificantes/farmacocinética , Animales , Femenino , Humanos , Intestinos , Hígado/metabolismo , Fase II de la Desintoxicación Metabólica , Modelos Animales , Oxidación-Reducción , Plasma/metabolismo , Embarazo , Ratas
7.
Inhal Toxicol ; 31(13-14): 468-483, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31992090

RESUMEN

Objective: To develop a physiologically based pharmacokinetic (PBPK) model for chloroprene in the mouse, rat and human, relying only on in vitro data to estimate tissue metabolism rates and partitioning, and to apply the model to calculate an inhalation unit risk (IUR) for chloroprene.Materials and methods: Female B6C3F1 mice were the most sensitive species/gender for lung tumors in the 2-year bioassay conducted with chloroprene. The PBPK model included tissue metabolism rate constants for chloroprene estimated from results of in vitro gas uptake studies using liver and lung microsomes. To assess the validity of the PBPK model, a 6-hr, nose-only chloroprene inhalation study was conducted with female B6C3F1 mice in which both chloroprene blood concentrations and ventilation rates were measured. The PBPK model was then used to predict dose measures - amounts of chloroprene metabolized in lungs per unit time - in mice and humans.Results: The mouse PBPK model accurately predicted in vivo pharmacokinetic data from the 6-hr, nose-only chloroprene inhalation study. The PBPK model was used to conduct a cancer risk assessment based on metabolism of chloroprene to reactive epoxides in the lung, the target tissue in mice. The IUR was over100-fold lower than the IUR from the EPA Integrated Risk Information System (IRIS), which was based on inhaled chloroprene concentration. The different result from the PBPK model risk assessment arises from use of the more relevant tissue dose metric, amount metabolized, rather than inhaled concentrationDiscussion and conclusions: The revised chloroprene PBPK model is based on the best available science, including new test animal in vivo validation, updated literature review and a Markov-Chain Monte Carlo analysis to assess parameter uncertainty. Relying on both mouse and human metabolism data also provides an important advancement in the use of quantitative in vitro to in vivo extrapolation (QIVIVE). Inclusion of the best available science is especially important when deriving a toxicity value based on species extrapolation for the potential carcinogenicity of a reactive metabolite.


Asunto(s)
Contaminantes Atmosféricos/farmacocinética , Cloropreno/farmacocinética , Exposición por Inhalación/efectos adversos , Pulmón/metabolismo , Modelos Biológicos , Contaminantes Atmosféricos/sangre , Contaminantes Atmosféricos/toxicidad , Animales , Cloropreno/sangre , Cloropreno/toxicidad , Femenino , Humanos , Exposición por Inhalación/análisis , Pulmón/efectos de los fármacos , Cadenas de Markov , Ratones , Método de Montecarlo , Pletismografía , Valor Predictivo de las Pruebas , Ratas , Medición de Riesgo , Especificidad de la Especie , Distribución Tisular
8.
Toxicol Appl Pharmacol ; 332: 149-158, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28392392

RESUMEN

Dichloromethane (DCM) is a lung and liver carcinogen in mice at inhalation exposures≥2000ppm. The modes of action (MOA) of these responses have been attributed to formation of genotoxic, reactive metabolite(s). Here, we examined gene expression in lung and liver from female B6C3F1 mice exposed to 0, 100, 500, 2000, 3000 and 4000ppm DCM for 90days. We also simulated dose measures - rates of DCM oxidation to carbon monoxide (CO) in lung and liver and expected blood carboxyhemoglobin (HbCO) time courses with a PBPK model inclusive of both conjugation and oxidation pathways. Expression of large numbers of genes was altered at 100ppm with maximal changes in the numbers occurring by 500 or 2000ppm. Most changes in genes common to the two tissues were related to cellular metabolism and circadian clock. At the lower concentrations, the changes in metabolism-related genes were discordant - up in liver and down in lung. These processes included organelle biogenesis, TCA cycle, and respiratory electron transport. Changes in circadian cycle genes - primarily transcription factors - showed strong concentration-related response at higher concentrations (Arntl, Npas2, and Clock were down-regulated; Cry2, Wee1, Bhlhe40, Per3, Nr1d1, Nr1d2 and Dbp) were up-regulated with similar directionality in both tissues. Overall, persistently elevated HbCO from DCM oxidation appears to cause extended periods of hypoxia, leading to altered circadian coupling to cellular metabolism. The dose response for altered circadian processes correlates with the cancer outcome. We found no evidence of changes in genes indicative of responses to cytotoxic, DNA-reactive metabolites.


Asunto(s)
Ritmo Circadiano , Hipoxia/genética , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Cloruro de Metileno/toxicidad , Transcriptoma , Animales , Carboxihemoglobina/genética , Carboxihemoglobina/metabolismo , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica , Hipoxia/inducido químicamente , Hipoxia/patología , Exposición por Inhalación/efectos adversos , Hígado/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos , Farmacocinética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Inhal Toxicol ; 28(11): 520-35, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27569524

RESUMEN

Benzo[a]pyrene (BaP) is a by-product of incomplete combustion of fossil fuels and plant/wood products, including tobacco. A physiologically based pharmacokinetic (PBPK) model for BaP for the rat was extended to simulate inhalation exposures to BaP in rats and humans including particle deposition and dissolution of absorbed BaP and renal elimination of 3-hydroxy benzo[a]pyrene (3-OH BaP) in humans. The clearance of particle-associated BaP from lung based on existing data in rats and dogs suggest that the process is bi-phasic. An initial rapid clearance was represented by BaP released from particles followed by a slower first-order clearance that follows particle kinetics. Parameter values for BaP-particle dissociation were estimated using inhalation data from isolated/ventilated/perfused rat lungs and optimized in the extended inhalation model using available rat data. Simulations of acute inhalation exposures in rats identified specific data needs including systemic elimination of BaP metabolites, diffusion-limited transfer rates of BaP from lung tissue to blood and the quantitative role of macrophage-mediated and ciliated clearance mechanisms. The updated BaP model provides very good prediction of the urinary 3-OH BaP concentrations and the relative difference between measured 3-OH BaP in nonsmokers versus smokers. This PBPK model for inhaled BaP is a preliminary tool for quantifying lung BaP dosimetry in rat and humans and was used to prioritize data needs that would provide significant model refinement and robust internal dosimetry capabilities.


Asunto(s)
Benzo(a)pireno/farmacocinética , Carcinógenos/farmacocinética , Pulmón/metabolismo , Modelos Biológicos , Material Particulado/farmacocinética , Administración por Inhalación , Administración Oral , Animales , Benzo(a)pireno/administración & dosificación , Benzopirenos/metabolismo , Carcinógenos/administración & dosificación , Humanos , Exposición por Inhalación , Material Particulado/administración & dosificación , Ratas
11.
Regul Toxicol Pharmacol ; 74 Suppl: S1-13, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26724268

RESUMEN

There are currently seven published physiologically based pharmacokinetic (PBPK) models describing aspects of the pharmacokinetics of octamethylcyclotetrasiloxane (D4) and decamethylcyclopentasiloxane (D5) for various exposure routes in rat and human. Each model addressed the biological and physico-chemical properties of D4 and D5 (highly lipophilic coupled with low blood: air partition coefficient and high liver clearance) that result in unique kinetic behaviors as well differences between D4 and D5. However, the proliferation of these models resulted in challenges for various risk assessment applications when needing to determine the optimum model for estimating dose metrics. To enhance the utility of these PBPK models for risk assessment, we integrated the suite of structures into one coherent model capable of simulating the entire set of existing data equally well as older more limited scope models. In this paper, we describe the steps required to develop this integrated model, the choice of physiological, partitioning and biochemical parameters for the model, and the concordance of the model behavior across key data sets. This integrated model is sufficiently robust to derive relevant dose metrics following individual or combined dermal and inhalation exposures of workers, consumer or the general population to D4 and D5 for route-to-route, interspecies and high to low dose extrapolations for risk assessment.


Asunto(s)
Modelos Biológicos , Siloxanos/farmacocinética , Tejido Adiposo/metabolismo , Animales , Femenino , Humanos , Exposición por Inhalación , Hígado/metabolismo , Masculino , Ratas , Medición de Riesgo , Piel/metabolismo , Distribución Tisular , Volatilización
12.
Crit Rev Toxicol ; 44(7): 600-17, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25068490

RESUMEN

Lipophilic persistent environmental chemicals (LPECs) have the potential to accumulate within a woman's body lipids over the course of many years prior to pregnancy, to partition into human milk, and to transfer to infants upon breastfeeding. As a result of this accumulation and partitioning, a breastfeeding infant's intake of these LPECs may be much greater than his/her mother's average daily exposure. Because the developmental period sets the stage for lifelong health, it is important to be able to accurately assess chemical exposures in early life. In many cases, current human health risk assessment methods do not account for differences between maternal and infant exposures to LPECs or for lifestage-specific effects of exposure to these chemicals. Because of their persistence and accumulation in body lipids and partitioning into breast milk, LPECs present unique challenges for each component of the human health risk assessment process, including hazard identification, dose-response assessment, and exposure assessment. Specific biological modeling approaches are available to support both dose-response and exposure assessment for lactational exposures to LPECs. Yet, lack of data limits the application of these approaches. The goal of this review is to outline the available approaches and to identify key issues that, if addressed, could improve efforts to apply these approaches to risk assessment of lactational exposure to these chemicals.


Asunto(s)
Contaminantes Ambientales/análisis , Exposición Materna , Leche Humana/química , Medición de Riesgo , Animales , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Modelos Teóricos , Método de Montecarlo , Embarazo , Ratas , Proyectos de Investigación
13.
Inhal Toxicol ; 26(6): 333-44, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24666369

RESUMEN

A PBPK model for naphthalene in the rat and human that incorporates a hybrid CFD-PBPK description of the upper respiratory tract was developed to support cross-species dosimetry comparisons of naphthalene concentrations and tissue normalized rate of metabolism in the nasal respiratory and olfactory epithelium, lung and liver. In vitro measurements of metabolic rates from microsomal incubations published for rat and monkey (surrogate for human) were scaled to the specific tissue based on the tissue microsomal content and volume of tissue. The model reproduces time courses for naphthalene blood concentrations from intravenous and inhalation exposures in rats and upper respiratory tract extraction data in both naïve rats and rats pre-treated to inhibit nasal metabolism. This naphthalene model was applied to estimate human equivalent inhalation concentrations (HECs) corresponding to several NOAELs or LOAELs for the non-cancer effects of naphthalene in rats. Two approaches for cross-species extrapolation were compared: (1) equivalence based on tissue naphthalene concentration and (2) equivalence based on amount metabolized per minute (normalized to tissue volume). At the NOAEL of 0.1 ppm, the regional gas dosimetry ratio (RGDR) based on naphthalene concentration was 0.18 for the dorsal olfactory region; however, the RGDR rises to 5.4 when based on the normalized amount metabolized due to the lower of expression of CYP isozymes in the nasal epithelium of primates and humans. The resulting HEC is 0.12 ppm (0.63 mg/m(3)) continuous exposure at the rat NOAEL of 0.1 ppm (6 h/day, 5 days/week).


Asunto(s)
Naftalenos/farmacocinética , Mucosa Nasal/metabolismo , Animales , Humanos , Hidrodinámica , Modelos Biológicos , Nivel sin Efectos Adversos Observados , Ratas , Especificidad de la Especie
14.
J Toxicol Environ Health A ; 76(1): 25-57, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23151209

RESUMEN

Perfluoroalkyl acid carboxylates and sulfonates (PFAA) have many consumer and industrial applications. Developmental toxicity studies in animals have raised concern about potential reproductive/developmental effects of perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS); however, in humans conflicting results have been reported for associations between maternal PFAA levels and these outcomes. Risk assessments and interpretation of available human data during gestation and lactation are hindered due to lack of a framework for understanding and estimating maternal, fetal, and neonatal pharmacokinetics (PK). Physiologically based pharmacokinetic (PBPK) models were developed for PFOA and PFOS for the gestation and lactation life stages in humans to understand how the physiological changes associated with development affect pharmacokinetics of these compounds in the mother, fetus, and infant. These models were derived from PBPK models for PFOA/PFOS that were previously developed for adult humans and rats during gestation and lactation and from existing human pregnancy and lactation models developed for other chemicals. The models simulated PFOA and PFOS concentrations in fetal, infant, and maternal plasma and milk, were compared to available data in humans, and also were used to estimate maternal exposure. The models reported here identified several research needs, which include (1) the identification of transporters involved in renal resorption to explain the multiyear half-lives of these compounds in humans, (2) factors affecting clearance of PFOA/PFOS during gestation and lactation, and (3) data to estimate clearance of PFOA/PFOS in infants. These models may help address concerns regarding possible adverse health effects due to PFOA/PFOS exposure in the fetus and infant and may be useful in comparing pharmacokinetics across life stages.


Asunto(s)
Ácidos Alcanesulfónicos/farmacocinética , Caprilatos/farmacocinética , Fluorocarburos/farmacocinética , Lactancia/metabolismo , Modelos Biológicos , Embarazo/metabolismo , Adulto , Animales , Simulación por Computador , Femenino , Haplorrinos , Humanos , Recién Nacido , Riñón/metabolismo , Exposición Materna , Intercambio Materno-Fetal , Unión Proteica , Ratas , Factores de Riesgo
15.
Toxicol Lett ; 375: 29-38, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36596351

RESUMEN

The most recent version of the octamethylcyclotetrasiloxane (D4) physiologically based pharmacokinetic (model) was developed using the available kinetic studies in male and female F344 rats. Additional data, which had not been included in the D4 model development, allowed for a more detailed assessment of the loss of D4 following long-term exposure in both SD and F344 rats. This new data demonstrated a deficiency in the published PBPK model predictions of terminal concentrations of D4 in plasma and fat 14 days after the end of exposures for 28-days, 6 h/day, where the model predictions were an order of magnitude lower than the data. To capture this time-point without altering the end-of-exposure peak concentrations in blood and fat required conversion of the one-way (liver to fat) mobile lipoprotein pool (MLP) into a bi-directional pool between liver and fat. Simulation of the D4 pharmacokinetics in the SD rat, as opposed to the F344 rat, also required a reduction of both fold induction of liver metabolism (KMAX: 5- to 2-fold) and maximal rate of metabolism (VMAXC: 5.0-1.54 mg/kg0.75). The revised MLP description was extended to the human D4 model using a parallelogram approach between rat and human MLP parameters to establish the parameters for the current model in the absence of similar long-term clearance data in the human. The revised human D4 model provided good fits to the human inhalation and dermal exposure studies while not appreciably altering cross-species dose metrics based on the free concentration of D4 in blood.


Asunto(s)
Exposición por Inhalación , Siloxanos , Ratas , Masculino , Femenino , Humanos , Animales , Ratas Endogámicas F344 , Exposición por Inhalación/efectos adversos , Siloxanos/toxicidad , Siloxanos/farmacocinética , Cinética , Ratas Sprague-Dawley , Modelos Biológicos , Lípidos
16.
Toxicol Sci ; 191(2): 212-226, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36453847

RESUMEN

In earlier physiologically based pharmacokinetic (PBPK) models for manganese (Mn), the kinetics of transport of Mn into and out of tissues were primarily driven by slow rates of association and dissociation of Mn with tissue binding sites. However, Mn is known to show rapidly reversible binding in tissues. An updated Mn model for primates, following similar work with rats, was developed that included rapid association/dissociation processes with tissue Mn-binding sites, accumulation of free Mn in tissues after saturation of these Mn-binding sites and rapid rates of entry into tissues. This alternative structure successfully described Mn kinetics in tissues in monkeys exposed to Mn via various routes including oral, inhalation, and intraperitoneal, subcutaneous, or intravenous injection and whole-body kinetics and tissue levels in humans. An important contribution of this effort is showing that the extension of the rate constants for binding and cellular uptake established in the monkey were also able to describe kinetic data from humans. With a consistent model structure for monkeys and humans, there is less need to rely on cadaver data and whole-body tracer studies alone to calibrate a human model. The increased biological relevance of the Mn model structure and parameters provides greater confidence in applying the Mn PBPK models to risk assessment. This model is also well-suited to explicitly incorporate emerging information on the role of transporters in tissue disposition, intestinal uptake, and hepatobiliary excretion of Mn.


Asunto(s)
Manganeso , Modelos Biológicos , Humanos , Ratas , Animales , Haplorrinos , Transporte Biológico , Administración por Inhalación
17.
Toxicol Sci ; 191(1): 15-24, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36409013

RESUMEN

Understanding the dose-response for formaldehyde-induced nasal cancer in rats is complicated by (1) the uneven distribution of inhaled formaldehyde across the interior surface of the nasal cavity and, (2) the presence of endogenous formaldehyde (endoF) in the nasal mucosa. In this work, we used computational fluid dynamics (CFD) modeling to predict flux of inhaled (exogenous) formaldehyde (exogF) from air into tissue at the specific locations where DNA adducts were measured. Experimental work has identified DNA-protein crosslink (DPX) adducts due to exogF and deoxyguanosine (DG) adducts due to both exogF and endoF. These adducts can be considered biomarkers of exposure for effects of endoF and exogF on DNA that may be part of the mechanism of tumor formation. We describe a computational model linking CFD-predicted flux of formaldehyde from air into tissue, and the intracellular production of endoF, with the formation of DPX and DG adducts. We assumed that, like exogF, endoF can produce DPX. The model accurately reproduces exogDPX, exogDG, and endoDG data after inhalation from 0.7 to 15 ppm. The dose-dependent concentrations of exogDPX and exogDG are predicted to exceed the concentrations of their endogenous counterparts at about 2 and 6 ppm exogF, respectively. At all concentrations examined, the concentrations of endoDPX and exogDPX were predicted to be at least 10-fold higher than that of their DG counterparts. The modeled dose-dependent concentrations of these adducts are suitable to be used together with data on the dose-dependence of cell proliferation to conduct quantitative modeling of formaldehyde-induced rat nasal carcinogenicity.


Asunto(s)
Aductos de ADN , ADN , Ratas , Animales , Ratas Endogámicas F344 , Mucosa Nasal , Formaldehído/toxicidad , Desoxiguanosina
18.
Front Pharmacol ; 14: 1088011, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36909196

RESUMEN

Introduction: A physiologically based pharmacokinetic (PBPK) model for 3-chloroallyl alcohol (3-CAA) was developed and used to evaluate the design of assays for the in vivo genotoxicity of 3-CAA. Methods: Model development was supported by read across from a published PBPK model for ethanol. Read across was motivated by the expectation that 3-CAA, which like ethanol is a primary alcohol, is metabolized largely by hepatic alcohol dehydrogenases. The PBPK model was used to evaluate how two metrics of tissue dosimetry, maximum blood concentration (Cmax; mg/L) and area under the curve (AUC; mg-hr/L) vary with dose of 3-CAA and with dose route (oral gavage, drinking water). Results: The model predicted that oral gavage results in a 6-fold higher Cmax than the same dose administered in drinking water, but in similar AUCs. Predicted Cmax provided the best correlation with severe toxicity (e.g., lethality) from 3-CAA, consistent with the production of a reactive metabolite. Therefore, drinking water administration can achieve higher sustained concentration without severe toxicity in vivo. Discussion: This evaluation is significant because cytotoxicity is a potential confounder of mutagenicity testing. The PBPK model can be used to ensure that studies meet OECD and USEPA test guidelines and that the highest dose used is not associated with severe toxicity. In addition, PBPK modeling provides assurance of target tissue (e.g., bone marrow) exposure even in the absence of laboratory data, by defining the relationship between applied dose and target tissue dose based on accepted principles of pharmacokinetics, relevant physiology and biochemistry of the dosed animals, and chemical-specific information.

19.
Crit Rev Toxicol ; 42(8): 633-52, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22667820

RESUMEN

The field of toxicology is currently undergoing a global paradigm shift to use of in vitro approaches for assessing the risks of chemicals and drugs in a more mechanistic and high throughput manner than current approaches relying primarily on in vivo testing. However, reliance on in vitro data entails a number of new challenges associated with translating the in vitro results to corresponding in vivo exposures. Physiologically based pharmacokinetic (PBPK) modeling provides an effective framework for conducting quantitative in vitro to in vivo extrapolation (QIVIVE). Their physiological structure facilitates the incorporation of in silico- and in vitro-derived chemical-specific parameters in order to predict in vivo absorption, distribution, metabolism and excretion. In particular, the combination of in silico- and in vitro parameter estimation with PBPK modeling can be used to predict the in vivo exposure conditions that would produce chemical concentrations in the target tissue equivalent to the concentrations at which effects were observed with in vitro assays of tissue/organ toxicity. This review describes the various elements of QIVIVE and highlights key aspects of the process, with an emphasis on extrapolation of in vitro metabolism data to predict in vivo clearance as the key element. Other important elements include characterization of free concentration in the toxicity assay and potential complications associated with intestinal absorption and renal clearance. Examples of successful QIVIVE approaches are described ranging from a simple steady-state approach that is suitable for a high throughput environment to more complicated approaches requiring full PBPK models.


Asunto(s)
Contaminantes Ambientales/toxicidad , Estudios de Evaluación como Asunto , Pruebas de Toxicidad/métodos , Animales , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/análisis , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Modelos Teóricos , Medición de Riesgo
20.
Inhal Toxicol ; 24(1): 1-26, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22188408

RESUMEN

The pharmacokinetic behavior of the majority of jet fuel constituents has not been previously described in the framework of a physiologically based pharmacokinetic (PBPK) model for inhalation exposure. Toxic effects have been reported in multiple organ systems, though exposure methods varied across studies, utilizing either vaporized or aerosolized fuels. The purpose of this work was to assess the pharmacokinetics of aerosolized and vaporized fuels, and develop a PBPK model capable of describing both types of exposures. To support model development, n-tetradecane and n-octane exposures were conducted at 89 mg/m(3) aerosol+vapor and 1000-5000 ppm vapor, respectively. Exposures to JP-8 and S-8 were conducted at ~900-1000 mg/m(3), and ~200 mg/m(3) to a 50:50 blend of both fuels. Sub-models were developed to assess the behavior of representative constituents and grouped unquantified constituents, termed "lumps", accounting for the remaining fuel mass. The sub-models were combined into the first PBPK model for petroleum and synthetic jet fuels. Inhalation of hydrocarbon vapors was described with simple gas-exchange assumptions for uptake and exhalation. For aerosol droplets systemic uptake occurred in the thoracic region. Visceral tissues were described using perfusion and diffusion-limited equations. The model described kinetics at multiple fuel concentrations, utilizing a chemical "lumping" strategy to estimate parameters for fractions of speciated and unspeciated hydrocarbons and gauge metabolic interactions. The model more accurately simulated aromatic and lower molecular weight (MW) n-alkanes than some higher MW chemicals. Metabolic interactions were more pronounced at high (~2700-1000 mg/m(3)) concentrations. This research represents the most detailed assessment of fuel pharmacokinetics to date.


Asunto(s)
Contaminantes Ocupacionales del Aire/farmacocinética , Hidrocarburos/farmacocinética , Modelos Biológicos , Tejido Adiposo/metabolismo , Administración por Inhalación , Contaminantes Ocupacionales del Aire/sangre , Animales , Encéfalo/metabolismo , Hidrocarburos/sangre , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Ratas , Ratas Endogámicas F344
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA