Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Comput Toxicol ; 222022 May.
Article En | MEDLINE | ID: mdl-35844258

Neurotoxicology is the study of adverse effects on the structure or function of the developing or mature adult nervous system following exposure to chemical, biological, or physical agents. The development of more informative alternative methods to assess developmental (DNT) and adult (NT) neurotoxicity induced by xenobiotics is critically needed. The use of such alternative methods including in silico approaches that predict DNT or NT from chemical structure (e.g., statistical-based and expert rule-based systems) is ideally based on a comprehensive understanding of the relevant biological mechanisms. This paper discusses known mechanisms alongside the current state of the art in DNT/NT testing. In silico approaches available today that support the assessment of neurotoxicity based on knowledge of chemical structure are reviewed, and a conceptual framework for the integration of in silico methods with experimental information is presented. Establishing this framework is essential for the development of protocols, namely standardized approaches, to ensure that assessments of NT and DNT based on chemical structures are generated in a transparent, consistent, and defendable manner.

2.
Top Cogn Sci ; 14(4): 860-872, 2022 10.
Article En | MEDLINE | ID: mdl-35634901

Most computational theories of cognition lack a representation of physiology. Understanding the cognitive effects of compounds present in the environment is important for explaining and predicting changes in cognition and behavior given exposure to toxins, pharmaceuticals, or the deprivation of critical compounds like oxygen. This research integrates physiologically based pharmacokinetic (PBPK) model predictions of caffeine concentrations in blood and tissues with ACT-R's fatigue module to predict the effects of caffeine on fatigue. Mapping between the PBPK model parameters and ACT-R model parameters is informed by the neurophysiological literature and established associations between ACT-R modules and brain regions. The results from three such parameter mappings are explored to explain observed data from sleep-deprived participants performing the psychomotor vigilance test with and without caffeine. Predicted caffeine concentrations in the brain are used to modulate procedural parameters in the fatigue module to explain caffeine's effects on multiple performance metrics.


Caffeine , Sleep Deprivation , Humans , Caffeine/pharmacology , Sleep Deprivation/psychology , Psychomotor Performance/physiology , Fatigue/psychology , Oxygen/pharmacology , Pharmaceutical Preparations
5.
Environ Health Perspect ; 129(4): 47013, 2021 04.
Article En | MEDLINE | ID: mdl-33929906

BACKGROUND: Humans are exposed to tens of thousands of chemical substances that need to be assessed for their potential toxicity. Acute systemic toxicity testing serves as the basis for regulatory hazard classification, labeling, and risk management. However, it is cost- and time-prohibitive to evaluate all new and existing chemicals using traditional rodent acute toxicity tests. In silico models built using existing data facilitate rapid acute toxicity predictions without using animals. OBJECTIVES: The U.S. Interagency Coordinating Committee on the Validation of Alternative Methods (ICCVAM) Acute Toxicity Workgroup organized an international collaboration to develop in silico models for predicting acute oral toxicity based on five different end points: Lethal Dose 50 (LD50 value, U.S. Environmental Protection Agency hazard (four) categories, Globally Harmonized System for Classification and Labeling hazard (five) categories, very toxic chemicals [LD50 (LD50≤50mg/kg)], and nontoxic chemicals (LD50>2,000mg/kg). METHODS: An acute oral toxicity data inventory for 11,992 chemicals was compiled, split into training and evaluation sets, and made available to 35 participating international research groups that submitted a total of 139 predictive models. Predictions that fell within the applicability domains of the submitted models were evaluated using external validation sets. These were then combined into consensus models to leverage strengths of individual approaches. RESULTS: The resulting consensus predictions, which leverage the collective strengths of each individual model, form the Collaborative Acute Toxicity Modeling Suite (CATMoS). CATMoS demonstrated high performance in terms of accuracy and robustness when compared with in vivo results. DISCUSSION: CATMoS is being evaluated by regulatory agencies for its utility and applicability as a potential replacement for in vivo rat acute oral toxicity studies. CATMoS predictions for more than 800,000 chemicals have been made available via the National Toxicology Program's Integrated Chemical Environment tools and data sets (ice.ntp.niehs.nih.gov). The models are also implemented in a free, standalone, open-source tool, OPERA, which allows predictions of new and untested chemicals to be made. https://doi.org/10.1289/EHP8495.


Government Agencies , Animals , Computer Simulation , Rats , Toxicity Tests, Acute , United States , United States Environmental Protection Agency
6.
Chem Res Toxicol ; 34(2): 345-354, 2021 02 15.
Article En | MEDLINE | ID: mdl-33206501

While exposure of humans to environmental hazards often occurs with complex chemical mixtures, the majority of existing toxicity data are for single compounds. The Globally Harmonized System of chemical classification (GHS) developed by the Organization for Economic Cooperation and Development uses the additivity formula for acute oral toxicity classification of mixtures, which is based on the acute toxicity estimate of individual ingredients. We evaluated the prediction of GHS category classifications for mixtures using toxicological data collected in the Integrated Chemical Environment (ICE) developed by the National Toxicology Program (United States Department of Health and Human Services). The ICE database contains in vivo acute oral toxicity data for ∼10,000 chemicals and for 582 mixtures with one or multiple active ingredients. By using the available experimental data for individual ingredients, we were able to calculate a GHS category for only half of the mixtures. To expand a set of components with acute oral toxicity data, we used the Collaborative Acute Toxicity Modeling Suite (CATMoS) implemented in the Open Structure-Activity/Property Relationship App to make predictions for active ingredients without available experimental data. As a result, we were able to make predictions for 503 mixtures/formulations with 72% accuracy for the GHS classification. For 186 mixtures with two or more active ingredients, the accuracy rate was 76%. The structure-based analysis of the misclassified mixtures did not reveal any specific structural features associated with the mispredictions. Our results demonstrate that CATMoS together with an additivity formula can be used to predict the GHS category for chemical mixtures.


Organic Chemicals/adverse effects , Toxicity Tests , Administration, Oral , Databases, Chemical , Humans , Organic Chemicals/administration & dosage , Structure-Activity Relationship
7.
J Expo Sci Environ Epidemiol ; 30(5): 898, 2020 09.
Article En | MEDLINE | ID: mdl-32647364

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
J Expo Sci Environ Epidemiol ; 30(5): 866-877, 2020 09.
Article En | MEDLINE | ID: mdl-32546826

Currently it is difficult to prospectively estimate human toxicokinetics (particularly for novel chemicals) in a high-throughput manner. The R software package httk has been developed, in part, to address this deficiency, and the aim of this investigation was to develop a generalized inhalation model for httk. The structure of the inhalation model was developed from two previously published physiologically based models from Jongeneelen and Berge (Ann Occup Hyg 55:841-864, 2011) and Clewell et al. (Toxicol Sci 63:160-172, 2001), while calculated physicochemical data was obtained from EPA's CompTox Chemicals Dashboard. In total, 142 exposure scenarios across 41 volatile organic chemicals were modeled and compared to published data. The slope of the regression line of best fit between log-transformed simulated and observed blood and exhaled breath concentrations was 0.46 with an r2 = 0.45 and a root mean square error (RMSE) of direct comparison between the log-transformed simulated and observed values of 1.11. Approximately 5.1% (n = 108) of the data points analyzed were >2 orders of magnitude different than expected. The volatile organic chemicals examined in this investigation represent small, generally lipophilic molecules. Ultimately this paper details a generalized inhalation component that integrates with the httk physiologically based toxicokinetic model to provide high-throughput estimates of inhalation chemical exposures.


Volatile Organic Compounds , Humans , Inhalation Exposure , Models, Biological , Risk Assessment , Toxicokinetics
9.
Mil Med ; 185(Suppl 1): 390-395, 2020 01 07.
Article En | MEDLINE | ID: mdl-32074307

BACKGROUND: Military aircrews' health status is critical to their mission readiness, as they perform physically and cognitively demanding tasks in nontraditional work environments. Research Objectives: Our objective is to develop a broad operational risk assessment framework and demonstrate its applicability to health risks to aircrews because of airborne chemical exposure, considering stressors such as heat and exertion. METHODS: Extrapolation of generic exposure standards to military aviation-specific conditions can include computation of risk-relevant internal dosimetry estimates by incorporating changes in breathing patterns and blood flow distribution because of aspects of the in-flight environment. We provide an example of the effects of exertion on peak blood concentrations of 1,2,4-trimethylbenzene computed using a physiologically based pharmacokinetic model. RESULTS: Existing published collections on the effects of flight-related stressors on breathing patterns and blood flow address only a limited number of stressors. Although data exist that can be used to develop operational exposure limits specific to military aircrew activities, efforts to integrate this information in specific chemical assessments have been limited. CONCLUSIONS: Efforts to develop operational exposure limits would benefit from guidance on how to make use of existing assessments and expanded databases of the impact of environmental stressors on adult human physiology.


Aircraft/instrumentation , Hazardous Substances/analysis , Occupational Exposure/analysis , Aerospace Medicine/methods , Aerospace Medicine/statistics & numerical data , Aircraft/statistics & numerical data , Benzene Derivatives/analysis , Benzene Derivatives/blood , Hazardous Substances/blood , Humans , Occupational Exposure/statistics & numerical data , Risk Assessment/methods , United States , United States Environmental Protection Agency/organization & administration , United States Environmental Protection Agency/statistics & numerical data
10.
Toxicol Appl Pharmacol ; 355: 28-42, 2018 09 15.
Article En | MEDLINE | ID: mdl-29932956

Acute exposure to high concentrations of H2S causes severe brain injury and long-term neurological disorders, but the mechanisms involved are not known. To better understand the cellular and molecular mechanisms involved in acute H2S-induced neurodegeneration we used a broad-spectrum proteomic analysis approach to identify key molecules and molecular pathways involved in the pathogenesis of acute H2S-induced neurotoxicity and neurodegeneration. Mice were subjected to acute inhalation exposure of up to750 ppm of H2S. H2S induced behavioral deficits and severe lesions including hemorrhage in the inferior colliculus (IC). The IC was microdissected for proteomic analysis. Tandem mass tags (TMT) liquid chromatography mass spectrometry (LC-MS/MS)-based quantitative proteomics was applied for protein identification and quantitation. LC-MS/MS identified 598, 562, and 546 altered proteomic changes at 2 h, and on days 2 and 4 post-H2S exposure, respectively. Of these, 77 proteomic changes were statistically significant at any of the 3 time points. Mass spectrometry data were subjected to Perseus 1.5.5.3 statistical analysis, and gene ontology heat map clustering. Expressions of several key molecules were verified to confirm H2S-dependent proteomics changes. Webgestalt pathway overrepresentation enrichment analysis with Panther engine revealed H2S exposure disrupted several biological processes including metabotropic glutamate receptor group 1 and inflammation mediated by chemokine and cytokine signaling pathways among others. Further analysis showed that energy metabolism, integrity of blood-brain barrier, hypoxic, and oxidative stress signaling pathways were also implicated. Collectively, this broad-spectrum proteomics data has provided important clues to follow up in future studies to further elucidate mechanisms of H2S-induced neurotoxicity.


Hydrogen Sulfide/toxicity , Inferior Colliculi/metabolism , Inferior Colliculi/pathology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Proteomics , Animals , Behavior, Animal/drug effects , Gene Expression/drug effects , Inhalation Exposure , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Seizures/chemically induced , Signal Transduction/drug effects
11.
PLoS One ; 11(3): e0149300, 2016.
Article En | MEDLINE | ID: mdl-26930410

The Institute of Medicine recommends that lactating women ingest 290 µg iodide/d and a nursing infant, less than two years of age, 110 µg/d. The World Health Organization, United Nations Children's Fund, and International Council for the Control of Iodine Deficiency Disorders recommend population maternal and infant urinary iodide concentrations ≥ 100 µg/L to ensure iodide sufficiency. For breast milk, researchers have proposed an iodide concentration range of 150-180 µg/L indicates iodide sufficiency for the mother and infant, however no national or international guidelines exist for breast milk iodine concentration. For the first time, a lactating woman and nursing infant biologically based model, from delivery to 90 days postpartum, was constructed to predict maternal and infant urinary iodide concentration, breast milk iodide concentration, the amount of iodide transferred in breast milk to the nursing infant each day and maternal and infant serum thyroid hormone kinetics. The maternal and infant models each consisted of three sub-models, iodide, thyroxine (T4), and triiodothyronine (T3). Using our model to simulate a maternal intake of 290 µg iodide/d, the average daily amount of iodide ingested by the nursing infant, after 4 days of life, gradually increased from 50 to 101 µg/day over 90 days postpartum. The predicted average lactating mother and infant urinary iodide concentrations were both in excess of 100 µg/L and the predicted average breast milk iodide concentration, 157 µg/L. The predicted serum thyroid hormones (T4, free T4 (fT4), and T3) in both the nursing infant and lactating mother were indicative of euthyroidism. The model was calibrated using serum thyroid hormone concentrations for lactating women from the United States and was successful in predicting serum T4 and fT4 levels (within a factor of two) for lactating women in other countries. T3 levels were adequately predicted. Infant serum thyroid hormone levels were adequately predicted for most data. For moderate iodide deficient conditions, where dietary iodide intake may range from 50 to 150 µg/d for the lactating mother, the model satisfactorily described the iodide measurements, although with some variation, in urine and breast milk. Predictions of serum thyroid hormones in moderately iodide deficient lactating women (50 µg/d) and nursing infants did not closely agree with mean reported serum thyroid hormone levels, however, predictions were usually within a factor of two. Excellent agreement between prediction and observation was obtained for a recent moderate iodide deficiency study in lactating women. Measurements included iodide levels in urine of infant and mother, iodide in breast milk, and serum thyroid hormone levels in infant and mother. A maternal iodide intake of 50 µg/d resulted in a predicted 29-32% reduction in serum T4 and fT4 in nursing infants, however the reduced serum levels of T4 and fT4 were within most of the published reference intervals for infant. This biologically based model is an important first step at integrating the rapid changes that occur in the thyroid system of the nursing newborn in order to predict adverse outcomes from exposure to thyroid acting chemicals, drugs, radioactive materials or iodine deficiency.


Breast Feeding , Dietary Supplements/analysis , Iodides/analysis , Iodides/urine , Lactation , Milk, Human/chemistry , Computer Simulation , Female , Humans , Infant, Newborn , Iodides/administration & dosage , Models, Biological , Thyroxine/blood , Triiodothyronine/blood
12.
Am J Disaster Med ; 11(2): 89-118, 2016.
Article En | MEDLINE | ID: mdl-28102531

The use of nerve agents such as sarin is as much a threat today as any other time in our history. The events in Syria in 2013 are proof of this. "The Obama administration asserted Sunday for the first time that the Syrian government used the nerve gas sarin to kill more than 1,400 people (August 21, 2013) in the world's gravest chemical weapons attack in 25 years." With these recent events clear in our mind, we must focus on the horrific nature of these chemical agents to devise a strategy that will enable first responders to counteract these insidious chemicals. This paper presents research on a physiologically based pharmacokinetic model to determine whether the current treatment protocol prescribed by the Center for Disease Control (CDC) and the US Army is effective in treating victims suffering from acute exposure symptoms. The model was used to determine what treatment should be used for victims suffering from mild exposure symptoms. The results indicate that the current CDC and US Army treatment is effective, but treatment with oxime therapy was not effective in alleviating symptoms of mild exposure. By applying these results, an effective treatment protocol was developed.


Antidotes/pharmacokinetics , Chemical Warfare Agents/pharmacokinetics , Organophosphate Poisoning/drug therapy , Oximes/pharmacokinetics , Sarin/pharmacokinetics , Antidotes/therapeutic use , Centers for Disease Control and Prevention, U.S. , Chemical Warfare Agents/poisoning , Humans , Models, Theoretical , Oximes/therapeutic use , Practice Guidelines as Topic , Pralidoxime Compounds/therapeutic use , Sarin/poisoning , Systems Analysis , United States
13.
Protein Pept Lett ; 22(10): 862-8, 2015.
Article En | MEDLINE | ID: mdl-26216268

Organophosphorus (OP) pesticides and nerve agents have been designed to inhibit the hydrolysis of the neurotransmitter acetylcholine by covalently binding to the active site serine of acetylcholinesterase while Alzheimer drugs and prophylactics, such as tacrine, are characterized by reversible binding. Historically, the guinea pig has been believed to be the best non-primate model for OP toxicology and medical countermeasure development because, similarly to humans, guinea pigs have low amounts of circulating OP metabolizing carboxylesterase. To explore the hypothesis that guinea pigs are the appropriate responder species for OP toxicology and medical countermeasure development, guinea pig acetylcholinesterase (gpAChE) was cloned into pENTR/D-TOPO, recombined into pT-Rex-DEST30 and expressed in Human Embryonic Kidney 293 cells. Recombinant gpAChE was purified to a specific activity of 800 U/mg using size exclusion and immobilized nickel affinity chromatography, with purity confirmed by gel electrophoresis. Ellman's assay was used to enzymatically characterize gpAChE, identifying a K(M) of 154±18.7 µmol L(-1) and a k(cat) of 4.79x10(4)±5.26x10(2) /sec. Apparent gpAChE IC50's for diisopropylfluorophosphate, dicrotophos, paraoxon, and an Alzheimer's drug, tacrine, were found to be 10.1±1.98, 337±108, 1.02±0.29 and 0.30±0.01 µmol L(-1), respectively. Apparent gpAChE inhibition constants for diisopropylfluorophosphate, dicrotophos, paraoxon, and tacrine were found to be 8.40±0.60, 4.50±0.30, 0.29±0.01 and 0.42±0.07 µmol L(-1), respectively. Lineweaver-Burk plots confirmed tacrine as a mixed inhibitor and paraoxon, dicrotophos and diisopropylfluorophosphate as irreversible non-competitive inhibitors. gpAChE bimolecular rate constants for diisopropylfluorophosphate, dicrotophos and paraoxon were found to be 1.44±0.33x10(4), 1.56±0.12x10(3) and 4.57± 0.23x10(5) L µmol(-1) min(-1), respectively. Although the blood levels of OP metabolizing carboxylesterases in the guinea pig are similar to the low levels in human blood, the gpAChE is different in its enzymology. Therefore, medical countermeasures against OP intoxication should be tested for efficacy with the recombinant form of gpAChE prior to initiating animal studies.


Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Organophosphorus Compounds/chemistry , Animals , Guinea Pigs , Humans , Recombinant Proteins/chemistry
14.
J Biomol Screen ; 20(9): 1142-9, 2015 Oct.
Article En | MEDLINE | ID: mdl-26078409

The current standard of care for treatment of organophosphate (OP) poisoning includes pretreatment with the weak reversible acetylcholinesterase (AChE) inhibitor pyridostigmine bromide. Because this drug is an AChE inhibitor, similar side effects exist as with OP poisoning. In an attempt to provide a therapeutic capable of mitigating AChE inhibition without such side effects, high-throughput screening was performed to identify a compound capable of increasing the catalytic activity of AChE. Herein, two such novel positive allosteric modulators (PAMs) of AChE are presented. These PAMs increase AChE activity threefold, but they fail to upshift the apparent IC50 of a variety of OPs. Further development and optimization of these compounds may lead to pre- and/or postexposure therapeutics with broad-spectrum efficacy against pesticide and nerve agent poisoning. In addition, they could be used to complement the current therapeutic standard of care to increase the activity of uninhibited AChE, potentially increasing the efficacy of current therapeutics in addition to altering the therapeutic window.


Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Enzyme Activators/chemistry , Organophosphate Poisoning/drug therapy , Allosteric Regulation , Animals , Drug Evaluation, Preclinical , Drug Synergism , High-Throughput Screening Assays , Humans , Mice
15.
Brain Res ; 1611: 101-13, 2015 Jun 22.
Article En | MEDLINE | ID: mdl-25842371

The brainstem represents a major tissue area affected by sarin organophosphate poisoning due to its function in respiratory and cardiovascular control. While the acute toxic effects of sarin on brainstem-related responses are relatively unknown, other brain areas e.g., cortex or cerebellum, have been studied more extensively. The study objective was to analyze the guinea pig brainstem toxicology response following sarin (2×LD50) exposure by proteome pathway analysis to gain insight into the complex regulatory mechanisms that lead to impairment of respiratory and cardiovascular control. Guinea pig exposure to sarin resulted in the typical acute behavior/physiology outcomes with death between 15 and 25min. In addition, brain and blood acetylcholinesterase activity was significantly reduced in the presence of sarin to 95%, and 89%, respectively, of control values. Isobaric-tagged (iTRAQ) liquid chromatography tandem mass spectrometry (LC-MS/MS) identified 198 total proteins of which 23% were upregulated, and 18% were downregulated following sarin exposure. Direct gene ontology (GO) analysis revealed a sarin-specific broad-spectrum proteomic profile including glutamate-mediated excitotoxicity, calcium overload, energy depletion responses, and compensatory carbohydrate metabolism, increases in ROS defense, DNA damage and chromatin remodeling, HSP response, targeted protein degradation (ubiquitination) and cell death response. With regards to the sarin-dependent effect on respiration, our study supports the potential interference of sarin with CO2/H(+) sensitive chemoreceptor neurons of the brainstem retrotrapezoid nucleus (RTN) that send excitatory glutamergic projections to the respiratory centers. In conclusion, this study gives insight into the brainstem broad-spectrum proteome following acute sarin exposure and the gained information will assist in the development of novel countermeasures.


Brain Stem/drug effects , Brain Stem/metabolism , Central Nervous System Agents/toxicity , Proteomics , Sarin/toxicity , Acetylcholinesterase/blood , Animals , Apoptosis/drug effects , Brain Stem/chemistry , Calcium-Binding Proteins/metabolism , Cholinesterase Inhibitors/toxicity , Chromatin Assembly and Disassembly/drug effects , DNA Damage/drug effects , Excitatory Amino Acid Transporter 1/metabolism , Female , Guinea Pigs , Heat-Shock Proteins/drug effects , Reactive Oxygen Species/metabolism
16.
Theor Biol Med Model ; 11: 42, 2014 Oct 02.
Article En | MEDLINE | ID: mdl-25273339

BACKGROUND: One of the leading causes of death and illness within the agriculture industry is through unintentionally ingesting or inhaling organophosphate pesticides. OP intoxication directly inhibits acetylcholinesterase, resulting in an excitatory signaling cascade leading to fasciculation, loss of control of bodily fluids, and seizures. METHODS: Our model was developed using a discrete, rules-based modeling approach in NetLogo. This model includes acetylcholinesterase, the nicotinic acetylcholine receptor responsible for signal transduction, a single release of acetylcholine, organophosphate inhibitors, and a theoretical novel medical countermeasure. We have parameterized the system considering the molecular reaction rate constants in an agent-based approach, as opposed to apparent macroscopic rates used in differential equation models. RESULTS: Our model demonstrates how the cholinergic crisis can be mitigated by therapeutic intervention with an acetylcholinesterase activator. Our model predicts signal rise rates and half-lives consistent with in vitro and in vivo data in the absence and presence of inhibitors. It also predicts the efficacy of theoretical countermeasures acting through three mechanisms: increasing catalytic turnover of acetylcholine, increasing acetylcholine binding affinity to the enzyme, and decreasing binding rates of inhibitors. CONCLUSION: We present a model of the neuromuscular junction confirming observed acetylcholine signaling data and suggesting that developing a countermeasure capable of reducing inhibitor binding, and not activator concentration, is the most important parameter for reducing organophosphate (OP) intoxication.


Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Models, Neurological , Neuromuscular Junction/drug effects , Animals , Anura , Organophosphate Poisoning/drug therapy , Reproducibility of Results
17.
PLoS One ; 9(9): e106101, 2014.
Article En | MEDLINE | ID: mdl-25184666

The widespread usage of methylphenidate (MPH) in the pediatric population has received considerable attention due to its potential effect on child development. For the first time a physiologically based pharmacokinetic (PBPK) model has been developed in juvenile and adult humans and nonhuman primates to quantitatively evaluate species- and age-dependent enantiomer specific pharmacokinetics of MPH and its primary metabolite ritalinic acid. The PBPK model was first calibrated in adult humans using in vitro enzyme kinetic data of MPH enantiomers, together with plasma and urine pharmacokinetic data with MPH in adult humans. Metabolism of MPH in the small intestine was assumed to account for the low oral bioavailability of MPH. Due to lack of information, model development for children and juvenile and adult nonhuman primates primarily relied on intra- and interspecies extrapolation using allometric scaling. The juvenile monkeys appear to metabolize MPH more rapidly than adult monkeys and humans, both adults and children. Model prediction performance is comparable between juvenile monkeys and children, with average root mean squared error values of 4.1 and 2.1, providing scientific basis for interspecies extrapolation of toxicity findings. Model estimated human equivalent doses in children that achieve similar internal dose metrics to those associated with pubertal delays in juvenile monkeys were found to be close to the therapeutic doses of MPH used in pediatric patients. This computational analysis suggests that continued pharmacovigilance assessment is prudent for the safe use of MPH.


Central Nervous System Stimulants/pharmacokinetics , Methylphenidate/analogs & derivatives , Methylphenidate/pharmacokinetics , Models, Statistical , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Administration, Oral , Adolescent , Adult , Animals , Biological Availability , Biotransformation , Brain/drug effects , Brain/metabolism , Central Nervous System Stimulants/blood , Child , Gonads/drug effects , Gonads/metabolism , Humans , Intestine, Small/drug effects , Intestine, Small/metabolism , Liver/drug effects , Liver/metabolism , Macaca mulatta , Male , Methylphenidate/blood , Models, Biological , Myocardium/metabolism
18.
J Pharm Sci ; 103(7): 2189-2198, 2014 Jul.
Article En | MEDLINE | ID: mdl-24832575

A mechanistic tissue composition model incorporating passive and active transport for the prediction of steady-state tissue:plasma partition coefficients (K(t:pl)) of chemicals in multiple mammalian species was used to assess interindividual and interspecies variability. This approach predicts K(t:pl) using chemical lipophilicity, pKa, phospholipid membrane binding, and the unbound plasma fraction, together with tissue fractions of water, neutral lipids, neutral and acidic phospholipids, proteins, and pH. Active transport K(t:pl) is predicted using Michaelis-Menten transport parameters. Species-specific biological properties were identified from 126 peer reviewed journal articles, listed in the Supporting Information, for mouse, rat, guinea pig, rabbit, beagle dog, pig, monkey, and human species. Means and coefficients of variation for biological properties were used in a Monte Carlo analysis to assess variability. The results show K(t:pl) interspecies variability for the brain, fat, heart, kidney, liver, lung, muscle, red blood cell, skin, and spleen, but uncertainty in the estimates obscured some differences. Compounds undergoing active transport are shown to have concentration-dependent K(t:pl). This tissue composition-based mechanistic model can be used to predict K(t:pl) for organic chemicals across eight species and 10 tissues, and can be an important component in drug development when scaling K(t:pl) from animal models to humans.


Models, Biological , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/metabolism , Animals , Biological Transport, Active , Dogs , Forecasting , Guinea Pigs , Haplorhini , Mice , Monte Carlo Method , Organ Specificity , Rabbits , Rats , Species Specificity , Swine , Tissue Distribution
19.
Arch Toxicol ; 87(4): 661-80, 2013 Apr.
Article En | MEDLINE | ID: mdl-23314320

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.


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
20.
Arch Toxicol ; 87(2): 281-9, 2013 Feb.
Article En | MEDLINE | ID: mdl-22990135

Organophosphates are a group of pesticides and chemical warfare nerve agents that inhibit acetylcholinesterase, the enzyme responsible for hydrolysis of the excitatory neurotransmitter acetylcholine. Numerous structural variants exist for this chemical class, and data regarding their toxicity can be difficult to obtain in a timely fashion. At the same time, their use as pesticides and military weapons is widespread, which presents a major concern and challenge in evaluating human toxicity. To address this concern, a quantitative structure-activity relationship (QSAR) was developed to predict pentavalent organophosphate oxon human acetylcholinesterase bimolecular rate constants. A database of 278 three-dimensional structures and their bimolecular rates was developed from 15 peer-reviewed publications. A database of simplified molecular input line entry notations and their respective acetylcholinesterase bimolecular rate constants are listed in Supplementary Material, Table I. The database was quite diverse, spanning 7 log units of activity. In order to describe their structure, 675 molecular descriptors were calculated using AMPAC 8.0 and CODESSA 2.7.10. Orthogonal projection to latent structures regression, bootstrap leave-random-many-out cross-validation and y-randomization were used to develop an externally validated consensus QSAR model. The domain of applicability was assessed by the William's plot. Six external compounds were outside the warning leverage indicating potential model extrapolation. A number of compounds had residuals >2 or <-2, indicating potential outliers or activity cliffs. The results show that the HOMO-LUMO energy gap contributed most significantly to the binding affinity. A mean training R (2) of 0.80, a mean test set R (2) of 0.76 and a consensus external test set R (2) of 0.66 were achieved using the QSAR. The training and external test set RMSE values were found to be 0.76 and 0.88. The results suggest that this QSAR model can be used in physiologically based pharmacokinetic/pharmacodynamic models of organophosphate toxicity to determine the rate of acetylcholinesterase inhibition.


Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/metabolism , Organophosphates/metabolism , Pesticides/metabolism , Quantitative Structure-Activity Relationship , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/toxicity , Computer Simulation , Humans , Organophosphates/chemistry , Organophosphates/toxicity , Pesticides/chemistry , Protein Binding
...