Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Food Chem Toxicol ; 187: 114597, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38492856

RESUMEN

CONTEXT: Transition to the use of recycled plastics raises an issue concerning safety assessment of Non Intentionally Added Substances (NIAS). To assess the mutagenic potential of the recycled polyethylene impurities and to evaluate the need to perform in vitro assays on recycled resins, this study lies in identifying existing NIAS associated with recycled Low/High Density Polyethylene and assessing the mutagenicity data-gaps by employing in silico tools. METHODS: Quantitative Structure-Activity Relationship (QSAR) models predicting Ames mutagenicity were selected from literature, then NIAS were run to 1/evaluate performances of each model, 2/apply a QSAR strategy on the NIAS molecular space and address data-gaps. RESULTS: Among the 165 NIAS identified, experimental Ames results were not found for 50 substances while the substances with experimental data were predominantly negatives. No individual model was able to predict all NIAS due to applicability domain limitations. Taking into account 1/calculated performances, 2/availability of applicability domain, 3/description of the Training Set, an Integrated Strategy was founded including Sarpy, Consensus and Protox to extend the applicability domain. CONCLUSION & PERSPECTIVES: Existing data and predictions generated by this strategy suggest a low mutagenic potential of NIAS. Further investigation is needed to explore other genotoxicity mechanisms.


Asunto(s)
Mutágenos , Relación Estructura-Actividad Cuantitativa , Mutágenos/toxicidad , Mutágenos/análisis , Pruebas de Mutagenicidad/métodos , Mutagénesis , Reciclaje , Simulación por Computador
2.
Chem Res Toxicol ; 35(8): 1383-1392, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35830964

RESUMEN

To reduce the number of animals and studies needed to fulfill the information requirements as required by Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) (EC no. 1907/2006), a read-across approach was used to support approximately 30 higher olefins. This study aimed to assess the absorption potential of higher olefins through the gut wall as the experimentally determined bioavailability which would strengthen the read-across hypothesis and justification, reducing the need for toxicity studies on all of the higher olefins. The absorption potential of a series of higher olefins (carbon range from 6 to 28, with five configurations of the double bond) was determined in the in vitro everted rat small intestinal sac model and subsequently ranked. In addition, in silico approaches were applied to predict the reactivity, lipophilicity, and permeability of higher olefins. In the in vitro model, everted sacs were incubated in "fed-state simulated small intestinal fluid" saturated with individual higher olefins. The sac contents were then collected, extracted, and analyzed for olefin content using gas chromatography with a flame ionization detector. The C6 to C10 molecules were readily absorbed into the intestinal sacs. Marked inter-compound differences were observed, with the amount of absorption generally decreasing with the increase in carbon number. Higher olefins with ≥C14 carbons were either not absorbed or very poorly absorbed. In the reactivity simulation study, the reactivity is well described by the position of the double bond rather than the number of carbon atoms. In the lipophilicity and permeability analysis, both parameter descriptors depend mainly on the number of carbon atoms and less on the position of the double bond. In conclusion, these new approach methodologies provide supporting information on any trends or breakpoints in intestinal uptake and a hazard matrix based on carbon number and position of the double bond. This matrix will further assist in the selection of substances for inclusion in the mammalian toxicity testing programme.


Asunto(s)
Alquenos , Absorción Intestinal , Animales , Carbono/metabolismo , Intestino Delgado , Mamíferos , Permeabilidad , Ratas
3.
Regul Toxicol Pharmacol ; 114: 104658, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32334037

RESUMEN

To facilitate the practical implementation of the guidance on the residue definition for dietary risk assessment, EFSA has organized an evaluation of applicability of existing in silico models for predicting the genotoxicity of pesticides and their metabolites, including literature survey, application of QSARs and development of Read Across methodologies. This paper summarizes the main results. For the Ames test, all (Q)SAR models generated statistically significant predictions, comparable with the experimental variability of the test. The reliability of the models for other assays/endpoints appears to be still far from optimality. Two new Read Across approaches were evaluated: Read Across was largely successful for predicting the Ames test results, but less for in vitro Chromosomal Aberrations. The worse results for non-Ames endpoints may be attributable to the several revisions of experimental protocols and evaluation criteria of results, that have made the databases qualitatively non-homogeneous and poorly suitable for modeling. Last, Parent/Metabolite structural differences (besides known Structural Alerts) that may, or may not cause changes in the Ames mutagenicity were identified and catalogued. The findings from this work are suitable for being integrated into Weight-of-Evidence and Tiered evaluation schemes. Areas needing further developments are pointed out.


Asunto(s)
Aberraciones Cromosómicas/efectos de los fármacos , Plaguicidas/toxicidad , Relación Estructura-Actividad Cuantitativa , Bases de Datos Factuales , Humanos , Modelos Moleculares , Estructura Molecular , Pruebas de Mutagenicidad , Plaguicidas/análisis , Plaguicidas/metabolismo , Medición de Riesgo
4.
Toxicology ; 387: 27-42, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28645577

RESUMEN

This paper reviews in silico models currently available for the prediction of skin permeability. A comprehensive discussion on the developed methods is presented, focusing on quantitative structure-permeability relationships. In addition, the mechanistic models and comparative studies that analyse different models are discussed. Limitations and strengths of the different approaches are highlighted together with the emergent issues and perspectives.


Asunto(s)
Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Absorción Cutánea , Piel/metabolismo , Administración Cutánea , Animales , Bases de Datos de Compuestos Químicos , Difusión , Humanos , Tamaño de la Partícula , Permeabilidad , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Relación Estructura-Actividad Cuantitativa , Piel/anatomía & histología , Piel/efectos de los fármacos , Absorción Cutánea/efectos de los fármacos
5.
Toxicology ; 392: 140-154, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-26836498

RESUMEN

The aim of this paper was to provide a proof of concept demonstrating that molecular modelling methodologies can be employed as a part of an integrated strategy to support toxicity prediction consistent with the mode of action/adverse outcome pathway (MoA/AOP) framework. To illustrate the role of molecular modelling in predictive toxicology, a case study was undertaken in which molecular modelling methodologies were employed to predict the activation of the peroxisome proliferator-activated nuclear receptor γ (PPARγ) as a potential molecular initiating event (MIE) for liver steatosis. A stepwise procedure combining different in silico approaches (virtual screening based on docking and pharmacophore filtering, and molecular field analysis) was developed to screen for PPARγ full agonists and to predict their transactivation activity (EC50). The performance metrics of the classification model to predict PPARγ full agonists were balanced accuracy=81%, sensitivity=85% and specificity=76%. The 3D QSAR model developed to predict EC50 of PPARγ full agonists had the following statistical parameters: q2cv=0.610, Nopt=7, SEPcv=0.505, r2pr=0.552. To support the linkage of PPARγ agonism predictions to prosteatotic potential, molecular modelling was combined with independently performed mechanistic mining of available in vivo toxicity data followed by ToxPrint chemotypes analysis. The approaches investigated demonstrated a potential to predict the MIE, to facilitate the process of MoA/AOP elaboration, to increase the scientific confidence in AOP, and to become a basis for 3D chemotype development.


Asunto(s)
Modelos Moleculares , PPAR gamma/metabolismo , Pruebas de Toxicidad/métodos , Animales , Sitios de Unión , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Bases de Datos de Proteínas , Hígado Graso/metabolismo , Hígado Graso/patología , Estudios de Factibilidad , Células HEK293 , Haplorrinos , Células Hep G2 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Estructura Molecular , PPAR gamma/genética , Unión Proteica , Relación Estructura-Actividad Cuantitativa , Reproducibilidad de los Resultados , Medición de Riesgo , Sensibilidad y Especificidad
6.
Methods Mol Biol ; 1425: 511-29, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27311479

RESUMEN

The toxicological assessment of DNA-reactive/mutagenic or clastogenic impurities plays an important role in the regulatory process for pharmaceuticals; in this context, in silico structure-based approaches are applied as primary tools for the evaluation of the mutagenic potential of the drug impurities. The general recommendations regarding such use of in silico methods are provided in the recent ICH M7 guideline stating that computational (in silico) toxicology assessment should be performed using two (Q)SAR prediction methodologies complementing each other: a statistical-based method and an expert rule-based method.Based on our consultant experience, we describe here a framework for in silico assessment of mutagenic potential of drug impurities. Two main applications of in silico methods are presented: (1) support and optimization of drug synthesis processes by providing early indication of potential genotoxic impurities and (2) regulatory evaluation of genotoxic potential of impurities in compliance with the ICH M7 guideline. Some critical case studies are also discussed.


Asunto(s)
Biología Computacional/métodos , Preparaciones Farmacéuticas/química , Simulación por Computador , Contaminación de Medicamentos , Guías como Asunto , Pruebas de Mutagenicidad/métodos , Preparaciones Farmacéuticas/análisis , Relación Estructura-Actividad Cuantitativa , Fenómenos Toxicológicos
7.
Int J Mol Sci ; 15(5): 7651-66, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24857909

RESUMEN

The comprehensive understanding of the precise mode of action and/or adverse outcome pathway (MoA/AOP) of chemicals has become a key step toward the development of a new generation of predictive toxicology tools. One of the challenges of this process is to test the feasibility of the molecular modelling approaches to explore key molecular initiating events (MIE) within the integrated strategy of MoA/AOP characterisation. The description of MoAs leading to toxicity and liver damage has been the focus of much interest. Growing evidence underlines liver PPARγ ligand-dependent activation as a key MIE in the elicitation of liver steatosis. Synthetic PPARγ full agonists are of special concern, since they may trigger a number of adverse effects not observed with partial agonists. In this study, molecular modelling was performed based on the PPARγ complexes with full agonists extracted from the Protein Data Bank. The receptor binding pocket was analysed, and the specific ligand-receptor interactions were identified for the most active ligands. A pharmacophore model was derived, and the most important pharmacophore features were outlined and characterised in relation to their specific role for PPARγ activation. The results are useful for the characterisation of the chemical space of PPARγ full agonists and could facilitate the development of preliminary filtering rules for the effective virtual ligand screening of compounds with PPARγ full agonistic activity.


Asunto(s)
Simulación de Dinámica Molecular , PPAR gamma/agonistas , Sitios de Unión , Bases de Datos de Proteínas , Hígado Graso/metabolismo , Hígado Graso/patología , Humanos , Ligandos , PPAR gamma/metabolismo , Unión Proteica , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína
8.
Toxicol Lett ; 220(1): 26-34, 2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-23566899

RESUMEN

In the present legislations, the use of methods alternative to animal testing is explicitly encouraged, to use animal testing only 'as a last resort' or to ban it. The use of alternative methods to replace kinetics or repeated dose in vivo tests is a challenging issue. We propose here a strategy based on in vitro tests and QSAR (Quantitative Structure Activity Relationship) models to calibrate a dose-response model predicting hepatotoxicity. The dose response consists in calibrating and coupling a PBPK (physiologically-based pharmacokinetic) model with a toxicodynamic model for cell viability. We applied our strategy to acetaminophen and compared three different ways to calibrate the PBPK model: only with in vitro and in silico methods, using rat data or using all available data including data on humans. Some estimates of kinetic parameters differed substantially among the three calibration processes, but, at the end, the three models were quite comparable in terms of liver toxicity predictions and close to the usual range of human overdose. For the model based on alternative methods, the good adequation with the two other models resulted from an overestimated renal elimination rate which compensated for the underestimation of the metabolism rate. Our study points out that toxicokinetics/toxicodynamics approaches, based on alternative methods and modelling only, can predict in vivo liver toxicity with accuracy comparable to in vivo methods.


Asunto(s)
Acetaminofén/farmacocinética , Acetaminofén/toxicidad , Analgésicos/farmacocinética , Analgésicos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Modelos Biológicos , Acetaminofén/química , Analgésicos/química , Alternativas a las Pruebas en Animales , Animales , Simulación por Computador , Relación Dosis-Respuesta a Droga , Predicción , Humanos , Masculino , Relación Estructura-Actividad Cuantitativa , Ratas , Ratas Sprague-Dawley
9.
Bioorg Med Chem ; 16(6): 3091-107, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18248816

RESUMEN

Human Rhinovirus (HRV) is the most important aetiologic agent of common cold in adults and children. HRV is a single-stranded, positive sense RNA virus and, despite the high level of conservation among different serotypes, sequence alignment of viral protease 3C with mammalian protease reveals no homology. Thus, protease 3C is an optimal target for the development of anti-HRV agents. In the present work we investigated the design, the synthesis and the development of new potential reversible inhibitors against HRV protease 3C. Docking studies on the crystallized structure of HRV2 protease 3C led us to the design and the synthesis of a series of 3,5 disubstituted benzamides able to act as analogues of the substrate. We also developed 1,3,5 trisubstituted benzamides where aromatic substitutions on the aryl ring led us to investigate the importance of pi-pi interaction on the stabilization of protease 3C-inhibitor complex. All structures were tested for enzymatic inhibition on HRV14 protease 3C. Results highlighted the inhibitory activity of compounds 13, 14, and 20 (91%, 81%, and 85% at 10 microM, respectively), with the latter exhibiting an ID(50) (dose that inhibits 50% of the viral cytopathic effect) on HRV-14=25 microg/ml.


Asunto(s)
Antivirales/química , Benzamidas/química , Benzamidas/farmacología , Resfriado Común/tratamiento farmacológico , Cisteína Endopeptidasas/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/química , Rhinovirus/enzimología , Proteínas Virales/efectos de los fármacos , Proteasas Virales 3C , Humanos , Concentración 50 Inhibidora , Relación Estructura-Actividad , Proteínas Virales/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...