Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paul M. Schlosser is active.

Publication


Featured researches published by Paul M. Schlosser.


Environmental Health Perspectives | 2014

Human Health Effects of Dichloromethane: Key Findings and Scientific Issues

Paul M. Schlosser; Ambuja S. Bale; Catherine F. Gibbons; Amina Wilkins; Glinda S. Cooper

Background: The U.S. Environmental Protection Agency (EPA) completed a toxicological review of tetrachloroethylene (perchloroethylene, PCE) in February 2012 in support of the Integrated Risk Information System (IRIS). Objectives: We reviewed key findings and scientific issues regarding the human health effects of PCE described in the U.S. EPA’s Toxicological Review of Tetrachloroethylene (Perchloroethylene). Methods: The updated assessment of PCE synthesized and characterized a substantial database of epidemiological, experimental animal, and mechanistic studies. Key scientific issues were addressed through modeling of PCE toxicokinetics, synthesis of evidence from neurological studies, and analyses of toxicokinetic, mechanistic, and other factors (tumor latency, severity, and background rate) in interpreting experimental animal cancer findings. Considerations in evaluating epidemiological studies included the quality (e.g., specificity) of the exposure assessment methods and other essential design features, and the potential for alternative explanations for observed associations (e.g., bias or confounding). Discussion: Toxicokinetic modeling aided in characterizing the complex metabolism and multiple metabolites that contribute to PCE toxicity. The exposure assessment approach—a key evaluation factor for epidemiological studies of bladder cancer, non-Hodgkin lymphoma, and multiple myeloma—provided suggestive evidence of carcinogenicity. Bioassay data provided conclusive evidence of carcinogenicity in experimental animals. Neurotoxicity was identified as a sensitive noncancer health effect, occurring at low exposures: a conclusion supported by multiple studies. Evidence was integrated from human, experimental animal, and mechanistic data sets in assessing adverse health effects of PCE. Conclusions: PCE is likely to be carcinogenic to humans. Neurotoxicity is a sensitive adverse health effect of PCE. Citation: Guyton KZ, Hogan KA, Scott CS, Cooper GS, Bale AS, Kopylev L, Barone S Jr, Makris SL, Glenn B, Subramaniam RP, Gwinn MR, Dzubow RC, Chiu WA. 2014. Human health effects of tetrachloroethylene: key findings and scientific issues. Environ Health Perspect 122:325–334; http://dx.doi.org/10.1289/ehp.1307359


Journal of Toxicology and Environmental Health-part B-critical Reviews | 2008

Approaches for Applications of Physiologically Based Pharmacokinetic Models in Risk Assessment

Chad M. Thompson; Babasaheb Sonawane; Hugh A. Barton; Robert S. DeWoskin; John C. Lipscomb; Paul M. Schlosser; Weihsueh A. Chiu; Kannan Krishnan

Physiologically based pharmacokinetic (PBPK) models are particularly useful for simulating exposures to environmental toxicants for which, unlike pharmaceuticals, there is often little or no human data available to estimate the internal dose of a putative toxic moiety in a target tissue or an appropriate surrogate. This article reviews the current state of knowledge and approaches for application of PBPK models in the process of deriving reference dose, reference concentration, and cancer risk estimates. Examples drawn from previous U.S. Environmental Protection Agency (EPA) risk assessments and human health risk assessments in peer-reviewed literature illustrate the ways and means of using PBPK models to quantify the pharmacokinetic component of the interspecies and intraspecies uncertainty factors as well as to conduct route to route, high dose to low dose and duration extrapolations. The choice of the appropriate dose metric is key to the use of the PBPK models for the various applications in risk assessment. Issues related to whether uncertainty factors are most appropriately applied before or after derivation of human equivalent dose (or concentration) continue to be explored. Scientific progress in the understanding of life stage and genetic differences in dosimetry and their impacts on variability in susceptibility, as well as ongoing development of analytical methods to characterize uncertainty in PBPK models, will make their use in risk assessment increasingly likely. As such, it is anticipated that when PBPK models are used to express adverse tissue responses in terms of the internal target tissue dose of the toxic moiety rather than the external concentration, the scientific basis of, and confidence in, risk assessments will be enhanced.


Toxicological Sciences | 2012

Physiologically Based Pharmacokinetic Model Use in Risk Assessment—Why Being Published Is Not Enough

Eva D. McLanahan; Hisham A. El-Masri; Lisa M. Sweeney; Leonid Kopylev; Harvey J. Clewell; John F. Wambaugh; Paul M. Schlosser

A panel of experts in physiologically based pharmacokinetic (PBPK) modeling and relevant quantitative methods was convened to describe and discuss model evaluation criteria, issues, and choices that arise in model application and computational tools for improving model quality for use in human health risk assessments (HHRAs). Although publication of a PBPK model in a peer-reviewed journal is a mark of good science, subsequent evaluation of published models and the supporting computer code is necessary for their consideration for use in HHRAs. Standardized model evaluation criteria and a thorough and efficient review process can reduce the number of review and revision iterations and hence the time needed to prepare a model for application. Efficient and consistent review also allows for rapid identification of needed model modifications to address HHRA-specific issues. This manuscript reports on the workshop where a process and criteria that were created for PBPK model review were discussed along with other issues related to model review and application in HHRA. Other issues include (1) model code availability, portability, and validity; (2) probabilistic (e.g., population-based) PBPK models and critical choices in parameter values to fully characterize population variability; and (3) approaches to integrating PBPK model outputs with other HHRA tools, including benchmark dose modeling. Two specific case study examples are provided to illustrate challenges that were encountered during the review and application process. By considering the frequent challenges encountered in the review and application of PBPK models during the model development phase, scientists may be better able to prepare their models for use in HHRAs.


Toxicological Sciences | 2013

Application of an Updated Physiologically Based Pharmacokinetic Model for Chloroform to Evaluate CYP2E1-Mediated Renal Toxicity in Rats and Mice

Alan F. Sasso; Paul M. Schlosser; Gregory L. Kedderis; Mary Beth Genter; John E. Snawder; Zheng Li; Susan Rieth; John C. Lipscomb

Physiologically based pharmacokinetic (PBPK) models are tools for interpreting toxicological data and extrapolating observations across species and route of exposure. Chloroform (CHCl(3)) is a chemical for which there are PBPK models available in different species and multiple sites of toxicity. Because chloroform induces toxic effects in the liver and kidneys via production of reactive metabolites, proper characterization of metabolism in these tissues is essential for risk assessment. Although hepatic metabolism of chloroform is adequately described by these models, there is higher uncertainty for renal metabolism due to a lack of species-specific data and direct measurements of renal metabolism. Furthermore, models typically fail to account for regional differences in metabolic capacity within the kidney. Mischaracterization of renal metabolism may have a negligible effect on systemic chloroform levels, but it is anticipated to have a significant impact on the estimated site-specific production of reactive metabolites. In this article, rate parameters for chloroform metabolism in the kidney are revised for rats, mice, and humans. New in vitro data were collected in mice and humans for this purpose and are presented here. The revised PBPK model is used to interpret data of chloroform-induced kidney toxicity in rats and mice exposed via inhalation and drinking water. Benchmark dose (BMD) modeling is used to characterize the dose-response relationship of kidney toxicity markers as a function of PBPK-derived internal kidney dose. Applying the PBPK model, it was also possible to characterize the dose response for a recent data set of rats exposed via multiple routes simultaneously. Consistent BMD modeling results were observed regardless of species or route of exposure.


Risk Analysis | 2014

The use of PBPK models to inform human health risk assessment: case study on perchlorate and radioiodide human lifestage models.

Eva D. McLanahan; Paul A. White; Lynn Flowers; Paul M. Schlosser

Physiologically-based pharmacokinetic (PBPK) models are often submitted to or selected by agencies, such as the U.S. Environmental Protection Agency (U.S. EPA) and Agency for Toxic Substances and Disease Registry, for consideration for application in human health risk assessment (HHRA). Recently, U.S. EPA evaluated the human PBPK models for perchlorate and radioiodide for their ability to estimate the relative sensitivity of perchlorate inhibition on thyroidal radioiodide uptake for various population groups and lifestages. The most well-defined mode of action of the environmental contaminant, perchlorate, is competitive inhibition of thyroidal iodide uptake by the sodium-iodide symporter (NIS). In this analysis, a six-step framework for PBPK model evaluation was followed, and with a few modifications, the models were determined to be suitable for use in HHRA to evaluate relative sensitivity among human lifestages. Relative sensitivity to perchlorate was determined by comparing the PBPK model predicted percent inhibition of thyroidal radioactive iodide uptake (RAIU) by perchlorate for different lifestages. A limited sensitivity analysis indicated that model parameters describing urinary excretion of perchlorate and iodide were particularly important in prediction of RAIU inhibition; therefore, a range of biologically plausible values available in the peer-reviewed literature was evaluated. Using the updated PBPK models, the greatest sensitivity to RAIU inhibition was predicted to be the near-term fetus (gestation week 40) compared to the average adult and other lifestages; however, when exposure factors were taken into account, newborns were found to be populations that need further evaluation and consideration in a risk assessment for perchlorate.


Toxicology and Applied Pharmacology | 2015

An evaluation of in vivo models for toxicokinetics of hexavalent chromium in the stomach.

Alan F. Sasso; Paul M. Schlosser

Hexavalent chromium (Cr6) is a drinking water contaminant that has been detected in most of the water systems throughout the United States. In 2-year drinking water bioassays, the National Toxicology Program (NTP) found clear evidence of carcinogenic activity in male and female rats and mice. Because reduction of Cr6 to trivalent chromium (Cr3) is an important detoxifying step in the gastrointestinal (GI) tract prior to systemic absorption, models have been developed to estimate the extent of reduction in humans and animals. The objective of this work was to use a revised model of ex vivo Cr6 reduction kinetics in gastric juice to analyze the potential reduction kinetics under in vivo conditions for mice, rats and humans. A published physiologically-based pharmacokinetic (PBPK) model was adapted to incorporate the new reduction model. This paper focuses on the toxicokinetics of Cr6 in the stomach compartment, where most of the extracellular Cr6 reduction is believed to occur in humans. Within the range of doses administered by the NTP bioassays, neither the original nor revised models predict saturation of stomach reducing capacity to occur in vivo if applying default parameters. However, both models still indicate that mice exhibit the lowest extent of reduction in the stomach, meaning that a higher percentage of the Cr6 dose may escape stomach reduction in that species. Similarly, both models predict that humans exhibit the highest extent of reduction at low doses.


Toxicology and Applied Pharmacology | 2011

Approaches to cancer assessment in EPA's Integrated Risk Information System

Martin W. Gehlhaus; Jeffrey S. Gift; Karen A. Hogan; Leonid Kopylev; Paul M. Schlosser; Abdel-Razak Kadry

The U.S. Environmental Protection Agencys (EPA) Integrated Risk Information System (IRIS) Program develops assessments of health effects that may result from chronic exposure to chemicals in the environment. The IRIS database contains more than 540 assessments. When supported by available data, IRIS assessments provide quantitative analyses of carcinogenic effects. Since publication of EPAs 2005 Guidelines for Carcinogen Risk Assessment, IRIS cancer assessments have implemented new approaches recommended in these guidelines and expanded the use of complex scientific methods to perform quantitative dose-response assessments. Two case studies of the application of the mode of action framework from the 2005 Cancer Guidelines are presented in this paper. The first is a case study of 1,2,3-trichloropropane, as an example of a chemical with a mutagenic mode of carcinogenic action thus warranting the application of age-dependent adjustment factors for early-life exposure; the second is a case study of ethylene glycol monobutyl ether, as an example of a chemical with a carcinogenic action consistent with a nonlinear extrapolation approach. The use of physiologically based pharmacokinetic (PBPK) modeling to quantify interindividual variability and account for human parameter uncertainty as part of a quantitative cancer assessment is illustrated using a case study involving probabilistic PBPK modeling for dichloromethane. We also discuss statistical issues in assessing trends and model fit for tumor dose-response data, analysis of the combined risk from multiple types of tumors, and application of life-table methods for using human data to derive cancer risk estimates. These issues reflect the complexity and challenges faced in assessing the carcinogenic risks from exposure to environmental chemicals, and provide a view of the current trends in IRIS carcinogenicity risk assessment.


Toxicology and Applied Pharmacology | 2014

A revised model of ex-vivo reduction of hexavalent chromium in human and rodent gastric juices

Paul M. Schlosser; Alan F. Sasso

Chronic oral exposure to hexavalent chromium (Cr-VI) in drinking water has been shown to induce tumors in the mouse gastrointestinal (GI) tract and rat oral cavity. The same is not true for trivalent chromium (Cr-III). Thus reduction of Cr-VI to Cr-III in gastric juices is considered a protective mechanism, and it has been suggested that the difference between the rate of reduction among mice, rats, and humans could explain or predict differences in sensitivity to Cr-VI. We evaluated previously published models of gastric reduction and believe that they do not fully describe the data on reduction as a function of Cr-VI concentration, time, and (in humans) pH. The previous models are parsimonious in assuming only a single reducing agent in rodents and describing pH-dependence using a simple function. We present a revised model that assumes three pools of reducing agents in rats and mice with pH-dependence based on known speciation chemistry. While the revised model uses more fitted parameters than the original model, they are adequately identifiable given the available data, and the fit of the revised model to the full range of data is shown to be significantly improved. Hence the revised model should provide better predictions of Cr-VI reduction when integrated into a corresponding PBPK model.


Toxicology Letters | 2014

Active and peripheral anionic sites of acetylcholinesterase have differential modulation effects on cell proliferation, adhesion and neuritogenesis in the NG108-15 cell line

Helen M. Campanha; Félix Carvalho; Paul M. Schlosser

The classical enzymatic role of acetylcholinesterase (AChE) is to terminate impulse transmission at cholinergic synapses through rapid hydrolysis of acetylcholine (ACh). Inactivation of this enzymes catalytic site is the primary mechanism of acute toxicity of OP insecticides (e.g. parathion, chlorpyrifos). There is now sufficient evidence to suggest that AChE has a neurotrophic function that may be altered by organophosphate (OP) exposure, resulting in defects of neuronal growth and development, though the clarification of the mechanisms involved require further in vitro investigation. In the present study, the mouse neuroblastoma×rat glioma hybrid NG108-15 cell line was used to investigate the differential effects between inhibition of the catalytic site and peripheral anionic site (PAS) of acetylcholinesterase (AChE) on cell adhesion, proliferation and neuritogenesis, in the presence and absence of human red blood cell (hRBC) AChE (ED3.1.1.7). AChE active-site inhibitor paraoxon (PO; 0.1-1.0μM), when added to NG108-15 cells grown on AChE-coated plates, had no effect on cell proliferation, but exerted a significant reduction in strongly adherent viable cells accompanied by mostly short process formations, with 18% of cells considered to be neuritogenic, similar to that observed on uncoated plates. In contrast, PO had no significant effect on cell adhesion and proliferation of NG108-15 cells on uncoated plates. The PAS-ligand thioflavin-T (Th-T; 0.5-25μM), however, decreased cell adhesion and proliferation, on both uncoated and ACh-E coated plates, with less magnitude on AChE-coated plates. Taken together, these results suggest that strong cell adherence and neuritogenesis are sensitive to PO in this cell culture model, with no impact on proliferation, in the presence of membrane bound AChE-coating, while there is no sensitivity to PO on uncoated plates. On the other hand, binding of Th-T directly to the PAS affects both cell adherence and proliferation, with less magnitude in the presence of membrane-bound AChE. The current study indicates that PO is deleterious in neural development during critical periods of strong cell adhesion and differentiation, interfering with AChE trophic function.


Journal of Applied Toxicology | 2016

Revision of the affinity constant for perchlorate binding to the sodium-iodide symporter based on in vitro and human in vivo data

Paul M. Schlosser

A series of previously published physiologically based pharmacokinetic (PBPK) models describe the effect of perchlorate on iodide uptake by the thyroid, with the mechanism being competitive inhibition of iodide transport by the sodium‐iodide symporter (NIS). Hence a key parameter of these models is the affinity of perchlorate for the NIS, characterized as the Michaelis–Menten kinetic constant, Km. However, when model predictions were compared to published results of a human study measuring radio‐iodide uptake (RAIU) inhibition after controlled perchlorate exposures, it was found to only fit the lowest exposure level and underpredicted RAIU inhibition at higher levels. Published in vitro data, in which perchlorate‐induced inhibition of iodide uptake via the NIS was measured, were re‐analyzed. Km for binding of perchlorate to the NIS originally derived from these data, 1.5 μm, had been obtained using Lineweaver–Burk plots, which allow for linear regression but invert the signal–noise of the data. Re‐fitting these data by non‐linear regression of the non‐inverted data yielded a 60% lower value for the Km, 0.59 μm. Substituting this value into the PBPK model for an average adult human significantly improved model agreement with the human RAIU data for exposures <100 μg kg−1 day−1. Thus, this lower Km value both fits the in vitro NIS kinetics and provides better predictions of human in vivo RAIU data. This change in Km increases the predicted sensitivity of humans to perchlorate over twofold for low‐level exposures. Published 2016. This article is a U.S. Government work and is in the public domain in the USA. Published 2016. This article is a U.S. Government work and is in the public domain in the USA.

Collaboration


Dive into the Paul M. Schlosser's collaboration.

Top Co-Authors

Avatar

Alan F. Sasso

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

John C. Lipscomb

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Babasaheb Sonawane

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Chad M. Thompson

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leonid Kopylev

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weihsueh A. Chiu

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge