1
|
Onasanwo AM, Mittapelly N, Shireman L, Vinden B, McNally K, Craig J, Bois FY. Using the Simcyp R Package for PBPK Simulation Workflows With the Simcyp Simulator. CPT Pharmacometrics Syst Pharmacol 2025; 14:853-863. [PMID: 40179011 PMCID: PMC12072214 DOI: 10.1002/psp4.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) modeling aims to understand how a drug is absorbed, distributed, metabolized, excreted, and acts in a human or animal body. The Simcyp Simulator is a well-known commercial PBPK/PD simulation software offering many options. It can, for example, use multiple compounds and population specification files to study the behavior of specific drug formulations in particular population groups. Such features can greatly speed up and optimize clinical research and drug development studies. On the other hand, the statistical coding language R offers benefits, such as easy scripting, automation, flexible statistical analyses, and visualization of results. These benefits can be applied to PBPK modeling. We describe here version 23.0.64 of an R software package that can run the Simcyp Simulator from R, changing its inputs and processing its outputs. We detail the implementation of two automated workflows for model development and verification. The first demonstrates the verification of a drug-drug interaction model for Atazanavir, an antiretroviral drug indicated for the treatment of HIV/AIDS. The second is applied to the virtual bioequivalence assessment of paliperidone palmitate long-acting injectable suspensions. We show how simulations that could take days otherwise can be executed, analyzed, and displayed in a matter of hours.
Collapse
Affiliation(s)
| | | | | | | | | | - James Craig
- CERTARA Data Science SoftwareRadnor, PACaliforniaUSA
| | | |
Collapse
|
2
|
Davidson-Fritz SE, Ring CL, Evans MV, Schacht CM, Chang X, Breen M, Honda GS, Kenyon E, Linakis MW, Meade A, Pearce RG, Sfeir MA, Sluka JP, Devito MJ, Wambaugh JF. Enabling transparent toxicokinetic modeling for public health risk assessment. PLoS One 2025; 20:e0321321. [PMID: 40238721 PMCID: PMC12002443 DOI: 10.1371/journal.pone.0321321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Toxicokinetic modeling describes the absorption, distribution, metabolism, and elimination of chemicals by the body. Chemical-specific in vivo toxicokinetic data is often unavailable for the thousands of chemicals in commerce. However, predictions from generalized toxicokinetic models allow for extrapolation from in vitro toxicological data, obtained via new approach methods (NAMs), to predict in vivo human health outcomes and provide key information on chemicals for public health risk assessment. The httk R package provides an open-source software tool containing a suite of generalized toxicokinetic models covering various exposure scenarios, a library of chemical-specific data from peer-reviewed high-throughput toxicokinetic (HTTK) studies, and other utility functions to parameterize and evaluate toxicokinetic models. Generalized HTTK models in httk use the open-source language MCSim to describe the compartmental and physiologically based toxicokinetics (PBTK). New HTTK models may be integrated into httk with a model description code file (C script generated via MCSim) and a model documentation file (R script). httk provides a series of functionalities such as model parameterization, in vivo-derived data for evaluating model predictions, unit conversion, Monte Carlo simulations for uncertainty propagation and biological variability, and other model utilities. Here, we describe in detail how to add new HTTK models into the httk package to leverage its pre-existing data and functionality. As a demonstration, we describe the integration of a gas inhalation PBTK model. The intention of httk is to provide a transparent, open-source tool for toxicokinetics, bioinformatics, and public health risk assessment that makes use of publicly available data on more than one thousand chemicals.
Collapse
Affiliation(s)
- Sarah E. Davidson-Fritz
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Cincinnati, Ohio, United States of America
| | - Caroline L. Ring
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Marina V. Evans
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Celia M. Schacht
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Xiaoqing Chang
- Inotiv, Research Triangle Park, North Carolina, United States of America
| | - Miyuki Breen
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Gregory S. Honda
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Elaina Kenyon
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | | | - Annabel Meade
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Robert G. Pearce
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Mark A. Sfeir
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - James P. Sluka
- Department of Intelligent Systems Engineering and Biocomplexity Institute, Indiana University, Bloomington, Indiana, United States of America
| | - Michael J. Devito
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - John F. Wambaugh
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
3
|
Tiryannik I, Heikkinen AT, Gardner I, Onasanwo A, Jamei M, Polasek TM, Rostami-Hodjegan A. Static Versus Dynamic Model Predictions of Competitive Inhibitory Metabolic Drug-Drug Interactions via Cytochromes P450: One Step Forward and Two Steps Backwards. Clin Pharmacokinet 2025; 64:155-170. [PMID: 39656410 PMCID: PMC11762507 DOI: 10.1007/s40262-024-01457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/26/2025]
Abstract
BACKGROUND Predicting metabolic drug-drug interactions (DDIs) via cytochrome P450 enzymes (CYP) is essential in drug development, but controversy has reemerged recently about whether in vitro-in vivo extrapolation (IVIVE) using static models can replace dynamic models for some regulatory filings and label recommendations. OBJECTIVE The aim of this study was to determine if static and dynamic models are equivalent for the quantitative prediction of metabolic DDIs arising from competitive CYP inhibition. METHODS Drug parameter spaces were varied to simulate 30,000 DDIs between hypothetical substrates and inhibitors of CYP3A4. Predicted area under the plasma concentration-time profile ratios for substrates (AUCr = AUC(presence of precipitant)/AUC(absence of precipitant)) were compared between dynamic simulations (Simcyp® V21) and corresponding static calculations, giving an inter-model discrepancy ratio (IMDR = AUCrdynamic/AUCrstatic). Dynamic simulations were conducted using a 'population' representative and a 'vulnerable patient' representative with maximal concentration (Cmax) or average steady-state concentration (Cavg,ss) as the inhibitor driver concentrations. IMDRs outside the interval 0.8-1.25 were defined as discrepancy between models. RESULTS The highest rate of IMDR <0.8 and IMDR >1.25 discrepancies in the 'population' representative was 85.9% and 3.1%, respectively, when using Cavg,ss as the inhibitor driver concentration. Using the 'vulnerable patient' representative showed the highest rate of IMDR >1.25 discrepancies at 37.8%. CONCLUSION Static models are not equivalent to dynamic models for predicting metabolic DDIs via competitive CYP inhibition across diverse drug parameter spaces, particularly for vulnerable patients. Caution is warranted in drug development if static IVIVE approaches are used alone to evaluate metabolic DDI risks.
Collapse
Affiliation(s)
- Ivan Tiryannik
- Certara Predictive Technologies (CPT), Sheffield, UK.
- Centre for Applied Pharmacokinetic Research (CAPKR), The University of Manchester, Manchester, UK.
| | | | - Iain Gardner
- Certara Predictive Technologies (CPT), Sheffield, UK
| | | | - Masoud Jamei
- Certara Predictive Technologies (CPT), Sheffield, UK
| | - Thomas M Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, Australia
- CMAX Clinical Research Pty Ltd, Adelaide, Australia
| | - Amin Rostami-Hodjegan
- Certara Predictive Technologies (CPT), Sheffield, UK
- Centre for Applied Pharmacokinetic Research (CAPKR), The University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Magel V, Blum J, Dolde X, Leisner H, Grillberger K, Khalidi H, Gardner I, Ecker GF, Pallocca G, Dreser N, Leist M. Inhibition of Neural Crest Cell Migration by Strobilurin Fungicides and Other Mitochondrial Toxicants. Cells 2024; 13:2057. [PMID: 39768149 PMCID: PMC11674305 DOI: 10.3390/cells13242057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Cell-based test methods with a phenotypic readout are frequently used for toxicity screening. However, guidance on how to validate the hits and how to integrate this information with other data for purposes of risk assessment is missing. We present here such a procedure and exemplify it with a case study on neural crest cell (NCC)-based developmental toxicity of picoxystrobin. A library of potential environmental toxicants was screened in the UKN2 assay, which simultaneously measures migration and cytotoxicity in NCC. Several strobilurin fungicides, known as inhibitors of the mitochondrial respiratory chain complex III, emerged as specific hits. From these, picoxystrobin was chosen to exemplify a roadmap leading from cell-based testing towards toxicological predictions. Following a stringent confirmatory testing, an adverse outcome pathway was developed to provide a testable toxicity hypothesis. Mechanistic studies showed that the oxygen consumption rate was inhibited at sub-µM picoxystrobin concentrations after a 24 h pre-exposure. Migration was inhibited in the 100 nM range, under assay conditions forcing cells to rely on mitochondria. Biokinetic modeling was used to predict intracellular concentrations. Assuming an oral intake of picoxystrobin, consistent with the acceptable daily intake level, physiologically based kinetic modeling suggested that brain concentrations of 0.1-1 µM may be reached. Using this broad array of hazard and toxicokinetics data, we calculated a margin of exposure ≥ 80 between the lowest in vitro point of departure and the highest predicted tissue concentration. Thus, our study exemplifies a hit follow-up strategy and contributes to paving the way to next-generation risk assessment.
Collapse
Affiliation(s)
- Viktoria Magel
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Jonathan Blum
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Xenia Dolde
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Heidrun Leisner
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Karin Grillberger
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Hiba Khalidi
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Iain Gardner
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Gerhard F. Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Giorgia Pallocca
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Nadine Dreser
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| |
Collapse
|
5
|
Geci R, Gadaleta D, de Lomana MG, Ortega-Vallbona R, Colombo E, Serrano-Candelas E, Paini A, Kuepfer L, Schaller S. Systematic evaluation of high-throughput PBK modelling strategies for the prediction of intravenous and oral pharmacokinetics in humans. Arch Toxicol 2024; 98:2659-2676. [PMID: 38722347 PMCID: PMC11272695 DOI: 10.1007/s00204-024-03764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/23/2024] [Indexed: 07/26/2024]
Abstract
Physiologically based kinetic (PBK) modelling offers a mechanistic basis for predicting the pharmaco-/toxicokinetics of compounds and thereby provides critical information for integrating toxicity and exposure data to replace animal testing with in vitro or in silico methods. However, traditional PBK modelling depends on animal and human data, which limits its usefulness for non-animal methods. To address this limitation, high-throughput PBK modelling aims to rely exclusively on in vitro and in silico data for model generation. Here, we evaluate a variety of in silico tools and different strategies to parameterise PBK models with input values from various sources in a high-throughput manner. We gather 2000 + publicly available human in vivo concentration-time profiles of 200 + compounds (IV and oral administration), as well as in silico, in vitro and in vivo determined compound-specific parameters required for the PBK modelling of these compounds. Then, we systematically evaluate all possible PBK model parametrisation strategies in PK-Sim and quantify their prediction accuracy against the collected in vivo concentration-time profiles. Our results show that even simple, generic high-throughput PBK modelling can provide accurate predictions of the pharmacokinetics of most compounds (87% of Cmax and 84% of AUC within tenfold). Nevertheless, we also observe major differences in prediction accuracies between the different parameterisation strategies, as well as between different compounds. Finally, we outline a strategy for high-throughput PBK modelling that relies exclusively on freely available tools. Our findings contribute to a more robust understanding of the reliability of high-throughput PBK modelling, which is essential to establish the confidence necessary for its utilisation in Next-Generation Risk Assessment.
Collapse
Affiliation(s)
- René Geci
- esqLABS GmbH, Saterland, Germany.
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany.
| | | | - Marina García de Lomana
- Machine Learning Research, Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | | | - Erika Colombo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | - Lars Kuepfer
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| | | |
Collapse
|
6
|
Pearce SM, Cross NA, Smith DP, Clench MR, Flint LE, Hamm G, Goodwin R, Langridge JI, Claude E, Cole LM. Multimodal Mass Spectrometry Imaging of an Osteosarcoma Multicellular Tumour Spheroid Model to Investigate Drug-Induced Response. Metabolites 2024; 14:315. [PMID: 38921450 PMCID: PMC11205347 DOI: 10.3390/metabo14060315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
A multimodal mass spectrometry imaging (MSI) approach was used to investigate the chemotherapy drug-induced response of a Multicellular Tumour Spheroid (MCTS) 3D cell culture model of osteosarcoma (OS). The work addresses the critical demand for enhanced translatable early drug discovery approaches by demonstrating a robust spatially resolved molecular distribution analysis in tumour models following chemotherapeutic intervention. Advanced high-resolution techniques were employed, including desorption electrospray ionisation (DESI) mass spectrometry imaging (MSI), to assess the interplay between metabolic and cellular pathways in response to chemotherapeutic intervention. Endogenous metabolite distributions of the human OS tumour models were complemented with subcellularly resolved protein localisation by the detection of metal-tagged antibodies using Imaging Mass Cytometry (IMC). The first application of matrix-assisted laser desorption ionization-immunohistochemistry (MALDI-IHC) of 3D cell culture models is reported here. Protein localisation and expression following an acute dosage of the chemotherapy drug doxorubicin demonstrated novel indications for mechanisms of region-specific tumour survival and cell-cycle-specific drug-induced responses. Previously unknown doxorubicin-induced metabolite upregulation was revealed by DESI-MSI of MCTSs, which may be used to inform mechanisms of chemotherapeutic resistance. The demonstration of specific tumour survival mechanisms that are characteristic of those reported for in vivo tumours has underscored the increasing value of this approach as a tool to investigate drug resistance.
Collapse
Affiliation(s)
- Sophie M. Pearce
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Neil A. Cross
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - David P. Smith
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Malcolm R. Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Lucy E. Flint
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Gregory Hamm
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Richard Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - James I. Langridge
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Emmanuelle Claude
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Laura M. Cole
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| |
Collapse
|
7
|
Braun G, Escher BI. Prioritization of mixtures of neurotoxic chemicals for biomonitoring using high-throughput toxicokinetics and mixture toxicity modeling. ENVIRONMENT INTERNATIONAL 2023; 171:107680. [PMID: 36502700 DOI: 10.1016/j.envint.2022.107680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Modern society continues to pollute the environment with larger quantities of chemicals that have also become more structurally and functionally diverse. Risk assessment of chemicals can hardly keep up with the sheer numbers that lead to complex mixtures of increasing chemical diversity including new chemicals, substitution products on top of still abundant legacy compounds. Fortunately, over the last years computational tools have helped us to identify and prioritize chemicals of concern. These include toxicokinetic models to predict exposure to chemicals as well as new approach methodologies such as in-vitro bioassays to address toxicodynamic effects. Combined, they allow for a prediction of mixtures and their respective effects and help overcome the lack of data we face for many chemicals. In this study we propose a high-throughput approach using experimental and predicted exposure, toxicokinetic and toxicodynamic data to simulate mixtures, to which a virtual population is exposed to and predict their mixture effects. The general workflow is adaptable for any type of toxicity, but we demonstrated its applicability with a case study on neurotoxicity. If no experimental data for neurotoxicity were available, we used baseline toxicity predictions as a surrogate. Baseline toxicity is the minimal toxicity any chemical has and might underestimate the true contribution to the mixture effect but many neurotoxicants are not by orders of magnitude more potent than baseline toxicity. Therefore, including baseline-toxic effects in mixture simulations yields a more realistic picture than excluding them in mixture simulations. This workflow did not only correctly identify and prioritize known chemicals of concern like benzothiazoles, organochlorine pesticides and plasticizers but we were also able to identify new potential neurotoxicants that we recommend to include in future biomonitoring studies and if found in humans, to also include in neurotoxicity screening.
Collapse
Affiliation(s)
- Georg Braun
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany.
| | - Beate I Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|