Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38645122

RESUMEN

Opioids are small-molecule agonists of µ-opioid receptor (µOR), while reversal agents such as naloxone are antagonists of µOR. Here we developed machine learning (ML) models to classify the intrinsic activities of ligands at the human µOR based on the SMILE strings and two-dimensional molecular descriptors. We first manually curated a database of 983 small molecules with measured E max values at the human µOR. Analysis of the chemical space allowed identification of dominant scaffolds and structurally similar agonists and antagonists. Decision tree models and directed message passing neural networks (MPNNs) were then trained to classify agonistic and antagonistic ligands. The hold-out test AUCs (areas under the receiver operator curves) of the extra-tree (ET) and MPNN models are 91.5±3.9% and 91.8± 4.4%, respectively. To overcome the challenge of small dataset, a student-teacher learning method called tri-training with disagreement was tested using an unlabeled dataset comprised of 15,816 ligands of human, mouse, or rat µOR, κOR, or δOR. We found that the tri-training scheme was able to increase the hold-out AUC of MPNN to as high as 95.7%. Our work demonstrates the feasibility of developing ML models to accurately predict the intrinsic activities of µOR ligands, even with limited data. We envisage potential applications of these models in evaluating uncharacterized substances for public safety risks and discovering new therapeutic agents to counteract opioid overdoses.

2.
bioRxiv ; 2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36945599

RESUMEN

The nation's opioid overdose deaths reached an all-time high in 2021. The majority of deaths are due to synthetic opioids represented by fentanyl. Naloxone, which is an FDA-approved reversal agent, antagonizes opioids through competitive binding at the mu-opioid receptor (mOR). Thus, knowledge of opioid's residence time is important for assessing the effectiveness of naloxone. Here we estimated the residence times of 15 fentanyl and 4 morphine analogs using metadynamics, and compared them with the most recent measurement of the opioid kinetic, dissociation, and naloxone inhibitory constants (Mann, Li et al, Clin. Pharmacol. Therapeut. 2022). Importantly, the microscopic simulations offered a glimpse at the common binding mechanism and molecular determinants of dissociation kinetics for fentanyl analogs. The insights inspired us to develop a machine learning (ML) approach to analyze the kinetic impact of fentanyl's substituents based on the interactions with mOR residues. This proof-of-concept approach is general; for example, it may be used to tune ligand residence times in computer-aided drug discovery.

3.
J Chem Inf Model ; 63(7): 2196-2206, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36977188

RESUMEN

The nation's opioid overdose deaths reached an all-time high in 2021. The majority of deaths are due to synthetic opioids represented by fentanyl. Naloxone, which is a FDA-approved reversal agent, antagonizes opioids through competitive binding at the µ-opioid receptor (mOR). Thus, knowledge of the opioid's residence time is important for assessing the effectiveness of naloxone. Here, we estimated the residence times (τ) of 15 fentanyl and 4 morphine analogs using metadynamics and compared them with the most recent measurement of the opioid kinetic, dissociation, and naloxone inhibitory constants (Mann et al. Clin. Pharmacol. Therapeut. 2022, 120, 1020-1232). Importantly, the microscopic simulations offered a glimpse at the common binding mechanism and molecular determinants of dissociation kinetics for fentanyl analogs. The insights inspired us to develop a machine learning approach to analyze the kinetic impact of fentanyl's substituents based on the interactions with mOR residues. This proof-of-concept approach is general; for example, it may be used to tune ligand residence times in computer-aided drug discovery.


Asunto(s)
Analgésicos Opioides , Naloxona , Analgésicos Opioides/farmacología , Naloxona/farmacología , Naloxona/metabolismo , Fentanilo/metabolismo , Fentanilo/farmacología , Morfina/química , Receptores Opioides mu/metabolismo , Antagonistas de Narcóticos
4.
Front Pharmacol ; 13: 1040838, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36339562

RESUMEN

Assessing drug permeability across the blood-brain barrier (BBB) is important when evaluating the abuse potential of new pharmaceuticals as well as developing novel therapeutics that target central nervous system disorders. One of the gold-standard in vivo methods for determining BBB permeability is rodent log BB; however, like most in vivo methods, it is time-consuming and expensive. In the present study, two statistical-based quantitative structure-activity relationship (QSAR) models were developed to predict BBB permeability of drugs based on their chemical structure. The in vivo BBB permeability data were harvested for 921 compounds from publicly available literature, non-proprietary drug approval packages, and University of Washington's Drug Interaction Database. The cross-validation performance statistics for the BBB models ranged from 82 to 85% in sensitivity and 80-83% in negative predictivity. Additionally, the performance of newly developed models was assessed using an external validation set comprised of 83 chemicals. Overall, performance of individual models ranged from 70 to 75% in sensitivity, 70-72% in negative predictivity, and 78-86% in coverage. The predictive performance was further improved to 93% in coverage by combining predictions across the two software programs. These new models can be rapidly deployed to predict blood brain barrier permeability of pharmaceutical candidates and reduce the use of experimental animals.

5.
Comput Toxicol ; 222022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35844258

RESUMEN

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.

6.
Front Med (Lausanne) ; 9: 1109541, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36743666

RESUMEN

The U.S. Food and Drug Administration (FDA) Division of Applied Regulatory Science (DARS) moves new science into the drug review process and addresses emergent regulatory and public health questions for the Agency. By forming interdisciplinary teams, DARS conducts mission-critical research to provide answers to scientific questions and solutions to regulatory challenges. Staffed by experts across the translational research spectrum, DARS forms synergies by pulling together scientists and experts from diverse backgrounds to collaborate in tackling some of the most complex challenges facing FDA. This includes (but is not limited to) assessing the systemic absorption of sunscreens, evaluating whether certain drugs can convert to carcinogens in people, studying drug interactions with opioids, optimizing opioid antagonist dosing in community settings, removing barriers to biosimilar and generic drug development, and advancing therapeutic development for rare diseases. FDA tasks DARS with wide ranging issues that encompass regulatory science; DARS, in turn, helps the Agency solve these challenges. The impact of DARS research is felt by patients, the pharmaceutical industry, and fellow regulators. This article reviews applied research projects and initiatives led by DARS and conducts a deeper dive into select examples illustrating the impactful work of the Division.

7.
Comput Toxicol ; 202021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35721273

RESUMEN

The kidneys, heart and lungs are vital organ systems evaluated as part of acute or chronic toxicity assessments. New methodologies are being developed to predict these adverse effects based on in vitro and in silico approaches. This paper reviews the current state of the art in predicting these organ toxicities. It outlines the biological basis, processes and endpoints for kidney toxicity, pulmonary toxicity, respiratory irritation and sensitization as well as functional and structural cardiac toxicities. The review also covers current experimental approaches, including off-target panels from secondary pharmacology batteries. Current in silico approaches for prediction of these effects and mechanisms are described as well as obstacles to the use of in silico methods. Ultimately, a commonly accepted protocol for performing such assessment would be a valuable resource to expand the use of such approaches across different regulatory and industrial applications. However, a number of factors impede their widespread deployment including a lack of a comprehensive mechanistic understanding, limited in vitro testing approaches and limited in vivo databases suitable for modeling, a limited understanding of how to incorporate absorption, distribution, metabolism, and excretion (ADME) considerations into the overall process, a lack of in silico models designed to predict a safe dose and an accepted framework for organizing the key characteristics of these organ toxicants.

8.
PLoS One ; 15(3): e0229646, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32126112

RESUMEN

Kratom is a botanical substance that is marketed and promoted in the US for pharmaceutical opioid indications despite having no US Food and Drug Administration approved uses. Kratom contains over forty alkaloids including two partial agonists at the mu opioid receptor, mitragynine and 7-hydroxymitragynine, that have been subjected to the FDA's scientific and medical evaluation. However, pharmacological and toxicological data for the remaining alkaloids are limited. Therefore, we applied the Public Health Assessment via Structural Evaluation (PHASE) protocol to generate in silico binding profiles for 25 kratom alkaloids to facilitate the risk evaluation of kratom. PHASE demonstrates that kratom alkaloids share structural features with controlled opioids, indicates that several alkaloids bind to the opioid, adrenergic, and serotonin receptors, and suggests that mitragynine and 7-hydroxymitragynine are the strongest binders at the mu opioid receptor. Subsequently, the in silico binding profiles of a subset of the alkaloids were experimentally verified at the opioid, adrenergic, and serotonin receptors using radioligand binding assays. The verified binding profiles demonstrate the ability of PHASE to identify potential safety signals and provide a tool for prioritizing experimental evaluation of high-risk compounds.


Asunto(s)
Mitragyna/química , Plantas Medicinales/química , Alcaloides de Triptamina Secologanina/química , Animales , Sitios de Unión , Células HEK293 , Humanos , Técnicas In Vitro , Simulación del Acoplamiento Molecular , Ensayo de Unión Radioligante , Receptores Adrenérgicos/efectos de los fármacos , Receptores Adrenérgicos/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Receptores de Serotonina/efectos de los fármacos , Receptores de Serotonina/metabolismo , Alcaloides de Triptamina Secologanina/farmacocinética , Alcaloides de Triptamina Secologanina/farmacología , Relación Estructura-Actividad
9.
Regul Toxicol Pharmacol ; 113: 104620, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32092371

RESUMEN

All drugs entering clinical trials are expected to undergo a series of in vitro and in vivo genotoxicity tests as outlined in the International Council on Harmonization (ICH) S2 (R1) guidance. Among the standard battery of genotoxicity tests used for pharmaceuticals, the in vivo micronucleus assay, which measures the frequency of micronucleated cells mostly from blood or bone marrow, is recommended for detecting clastogens and aneugens. (Quantitative) structure-activity relationship [(Q)SAR] models may be used as early screening tools by pharmaceutical companies to assess genetic toxicity risk during drug candidate selection. Models can also provide decision support information during regulatory review as part of the weight-of-evidence when experimental data are insufficient. In the present study, two commercial (Q)SAR platforms were used to construct in vivo micronucleus models from a recently enhanced in-house database of non-proprietary study findings in mice. Cross-validated performance statistics for the new models showed sensitivity of up to 74% and negative predictivity of up to 86%. In addition, the models demonstrated cross-validated specificity of up to 77% and coverage of up to 94%. These new models will provide more reliable predictions and offer an investigational approach for drug safety assessment with regards to identifying potentially genotoxic compounds.


Asunto(s)
Desarrollo de Medicamentos , Preparaciones Farmacéuticas/química , Relación Estructura-Actividad Cuantitativa , Animales , Aberraciones Cromosómicas , Bases de Datos Factuales , Ratones , Pruebas de Micronúcleos , Modelos Moleculares , Estructura Molecular , Pruebas de Mutagenicidad
10.
Regul Toxicol Pharmacol ; 109: 104488, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31586682

RESUMEN

The International Council on Harmonisation (ICH) M7(R1) guideline describes the use of complementary (quantitative) structure-activity relationship ((Q)SAR) models to assess the mutagenic potential of drug impurities in new and generic drugs. Historically, the CASE Ultra and Leadscope software platforms used two different statistical-based models to predict mutations at G-C (guanine-cytosine) and A-T (adenine-thymine) sites, to comprehensively assess bacterial mutagenesis. In the present study, composite bacterial mutagenicity models covering multiple mutation types were developed. These new models contain more than double the number of chemicals (n = 9,254 and n = 13,514) than the corresponding non-composite models and show better toxicophore coverage. Additionally, the use of a single composite bacterial mutagenicity model simplifies impurity analysis in an ICH M7 (Q)SAR workflow by reducing the number of model outputs requiring review. An external validation set of 388 drug impurities representing proprietary pharmaceutical chemical space showed performance statistics ranging from of 66-82% in sensitivity, 91-95% in negative predictivity and 96% in coverage. This effort represents a major enhancement to these (Q)SAR models and their use under ICH M7(R1), leading to improved patient safety through greater predictive accuracy, applicability, and efficiency when assessing the bacterial mutagenic potential of drug impurities.


Asunto(s)
Contaminación de Medicamentos/prevención & control , Mutagénesis/efectos de los fármacos , Pruebas de Mutagenicidad/normas , Mutágenos/toxicidad , Relación Estructura-Actividad Cuantitativa , Bacterias/efectos de los fármacos , Bacterias/genética , Simulación por Computador/normas , Exactitud de los Datos , Análisis de Datos , Bases de Datos Factuales , Conjuntos de Datos como Asunto , Humanos , Pruebas de Mutagenicidad/métodos , Mutágenos/química , Seguridad del Paciente , Proyectos de Investigación , Toxicología/métodos , Toxicología/normas , Flujo de Trabajo
11.
Clin Pharmacol Ther ; 106(1): 116-122, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30957872

RESUMEN

The US Food and Drug Administration's Center for Drug Evaluation and Research (CDER) developed an investigational Public Health Assessment via Structural Evaluation (PHASE) methodology to provide a structure-based evaluation of a newly identified opioid's risk to public safety. PHASE utilizes molecular structure to predict biological function. First, a similarity metric quantifies the structural similarity of a new drug relative to drugs currently controlled in the Controlled Substances Act (CSA). Next, software predictions provide the primary and secondary biological targets of the new drug. Finally, molecular docking estimates the binding affinity at the identified biological targets. The multicomponent computational approach coupled with expert review provides a rapid, systematic evaluation of a new drug in the absence of in vitro or in vivo data. The information provided by PHASE has the potential to inform law enforcement agencies with vital information regarding newly emerging illicit opioids.


Asunto(s)
Analgésicos Opioides/química , Sustancias Controladas/química , Control de Medicamentos y Narcóticos/organización & administración , Simulación del Acoplamiento Molecular/métodos , United States Food and Drug Administration/organización & administración , Simulación por Computador , Diseño de Fármacos , Fentanilo/química , Humanos , Salud Pública , Relación Estructura-Actividad , Estados Unidos
12.
Mutagenesis ; 34(1): 67-82, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30189015

RESUMEN

(Quantitative) structure-activity relationship or (Q)SAR predictions of DNA-reactive mutagenicity are important to support both the design of new chemicals and the assessment of impurities, degradants, metabolites, extractables and leachables, as well as existing chemicals. Aromatic N-oxides represent a class of compounds that are often considered alerting for mutagenicity yet the scientific rationale of this structural alert is not clear and has been questioned. Because aromatic N-oxide-containing compounds may be encountered as impurities, degradants and metabolites, it is important to accurately predict mutagenicity of this chemical class. This article analysed a series of publicly available aromatic N-oxide data in search of supporting information. The article also used a previously developed structure-activity relationship (SAR) fingerprint methodology where a series of aromatic N-oxide substructures was generated and matched against public and proprietary databases, including pharmaceutical data. An assessment of the number of mutagenic and non-mutagenic compounds matching each substructure across all sources was used to understand whether the general class or any specific subclasses appear to lead to mutagenicity. This analysis resulted in a downgrade of the general aromatic N-oxide alert. However, it was determined there were enough public and proprietary data to assign the quindioxin and related chemicals as well as benzo[c][1,2,5]oxadiazole 1-oxide subclasses as alerts. The overall results of this analysis were incorporated into Leadscope's expert-rule-based model to enhance its predictive accuracy.


Asunto(s)
Óxidos N-Cíclicos/química , Daño del ADN/efectos de los fármacos , Mutágenos/química , Relación Estructura-Actividad Cuantitativa , Óxidos N-Cíclicos/toxicidad , Mutagénesis/efectos de los fármacos , Pruebas de Mutagenicidad , Mutágenos/toxicidad
13.
Regul Toxicol Pharmacol ; 102: 53-64, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30562600

RESUMEN

The International Council for Harmonization (ICH) M7 guideline describes a hazard assessment process for impurities that have the potential to be present in a drug substance or drug product. In the absence of adequate experimental bacterial mutagenicity data, (Q)SAR analysis may be used as a test to predict impurities' DNA reactive (mutagenic) potential. However, in certain situations, (Q)SAR software is unable to generate a positive or negative prediction either because of conflicting information or because the impurity is outside the applicability domain of the model. Such results present challenges in generating an overall mutagenicity prediction and highlight the importance of performing a thorough expert review. The following paper reviews pharmaceutical and regulatory experiences handling such situations. The paper also presents an analysis of proprietary data to help understand the likelihood of misclassifying a mutagenic impurity as non-mutagenic based on different combinations of (Q)SAR results. This information may be taken into consideration when supporting the (Q)SAR results with an expert review, especially when out-of-domain results are generated during a (Q)SAR evaluation.


Asunto(s)
Contaminación de Medicamentos , Guías como Asunto , Mutágenos/clasificación , Relación Estructura-Actividad Cuantitativa , Industria Farmacéutica , Agencias Gubernamentales , Mutágenos/toxicidad , Medición de Riesgo
14.
Regul Toxicol Pharmacol ; 99: 274-288, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30278198

RESUMEN

In drug development, genetic toxicology studies are conducted using in vitro and in vivo assays to identify potential mutagenic and clastogenic effects, as outlined in the International Council for Harmonisation (ICH) S2 regulatory guideline. (Quantitative) structure-activity relationship ((Q)SAR) models that predict assay outcomes can be used as an early screen to prioritize pharmaceutical candidates, or later during product development to evaluate safety when experimental data are unavailable or inconclusive. In the current study, two commercial QSAR platforms were used to build models for in vitro chromosomal aberrations in Chinese hamster lung (CHL) and Chinese hamster ovary (CHO) cells. Cross-validated CHL model predictive performance showed sensitivity of 80 and 82%, and negative predictivity of 75 and 76% based on 875 training set compounds. For CHO, sensitivity of 61 and 67% and negative predictivity of 68 and 74% was achieved based on 817 training set compounds. The predictive performance of structural alerts in a commercial expert rule-based SAR software was also investigated and showed positive predictivity of 48-100% for selected alerts. Case studies examining incorrectly-predicted compounds, non-DNA-reactive clastogens, and recently-approved pharmaceuticals are presented, exploring how an investigational approach using similarity searching and expert knowledge can improve upon individual (Q)SAR predictions of the clastogenicity of drugs.


Asunto(s)
Aberraciones Cromosómicas/inducido químicamente , Mutágenos/efectos adversos , Mutágenos/química , Animales , Células CHO , Línea Celular , Simulación por Computador , Cricetinae , Cricetulus , Contaminación de Medicamentos , Mutagénesis/efectos de los fármacos , Pruebas de Mutagenicidad/métodos , Relación Estructura-Actividad Cuantitativa , Ratas , Programas Informáticos
15.
PLoS One ; 13(5): e0197734, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29795628

RESUMEN

Opioids represent a highly-abused and highly potent class of drugs that have become a significant threat to public safety. Often there are little to no pharmacological and toxicological data available for new, illicitly used and abused opioids, and this has resulted in a growing number of serious adverse events, including death. The large influx of new synthetic opioids permeating the street-drug market, including fentanyl and fentanyl analogs, has generated the need for a fast and effective method to evaluate the risk a substance poses to public safety. In response, the US FDA's Center for Drug Evaluation and Research (CDER) has developed a rapidly-deployable, multi-pronged computational approach to assess a drug's risk to public health. A key component of this approach is a molecular docking model to predict the binding affinity of biologically uncharacterized fentanyl analogs to the mu opioid receptor. The model was validated by correlating the docking scores of structurally diverse opioids with experimentally determined binding affinities. Fentanyl derivatives with sub-nanomolar binding affinity at the mu receptor (e.g. carfentanil and lofentanil) have significantly lower binding scores, while less potent fentanyl derivatives have increased binding scores. The strong correlation between the binding scores and the experimental binding affinities suggests that this approach can be used to accurately predict the binding strength of newly identified fentanyl analogs at the mu receptor in the absence of in vitro data and may assist in the temporary scheduling of those substances that pose a risk to public safety.


Asunto(s)
Fentanilo/metabolismo , Simulación del Acoplamiento Molecular , Receptores Opioides mu/metabolismo , Sitios de Unión , Fentanilo/análogos & derivados , Fentanilo/química , Humanos , Cinética , Unión Proteica , Estructura Terciaria de Proteína , Receptores Opioides mu/química , Termodinámica
16.
Regul Toxicol Pharmacol ; 77: 13-24, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26877192

RESUMEN

The ICH M7 guideline describes a consistent approach to identify, categorize, and control DNA reactive, mutagenic, impurities in pharmaceutical products to limit the potential carcinogenic risk related to such impurities. This paper outlines a series of principles and procedures to consider when generating (Q)SAR assessments aligned with the ICH M7 guideline to be included in a regulatory submission. In the absence of adequate experimental data, the results from two complementary (Q)SAR methodologies may be combined to support an initial hazard classification. This may be followed by an assessment of additional information that serves as the basis for an expert review to support or refute the predictions. This paper elucidates scenarios where additional expert knowledge may be beneficial, what such an expert review may contain, and how the results and accompanying considerations may be documented. Furthermore, the use of these principles and procedures to yield a consistent and robust (Q)SAR-based argument to support impurity qualification for regulatory purposes is described in this manuscript.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Daño del ADN , Minería de Datos/métodos , Mutagénesis , Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Toxicología/métodos , Animales , Pruebas de Carcinogenicidad/normas , Simulación por Computador , Bases de Datos Factuales , Adhesión a Directriz , Guías como Asunto , Humanos , Modelos Moleculares , Estructura Molecular , Pruebas de Mutagenicidad/normas , Mutágenos/química , Mutágenos/clasificación , Formulación de Políticas , Relación Estructura-Actividad Cuantitativa , Medición de Riesgo , Toxicología/legislación & jurisprudencia , Toxicología/normas
17.
Regul Toxicol Pharmacol ; 77: 1-12, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26879463

RESUMEN

Statistical-based and expert rule-based models built using public domain mutagenicity knowledge and data are routinely used for computational (Q)SAR assessments of pharmaceutical impurities in line with the approach recommended in the ICH M7 guideline. Knowledge from proprietary corporate mutagenicity databases could be used to increase the predictive performance for selected chemical classes as well as expand the applicability domain of these (Q)SAR models. This paper outlines a mechanism for sharing knowledge without the release of proprietary data. Primary aromatic amine mutagenicity was selected as a case study because this chemical class is often encountered in pharmaceutical impurity analysis and mutagenicity of aromatic amines is currently difficult to predict. As part of this analysis, a series of aromatic amine substructures were defined and the number of mutagenic and non-mutagenic examples for each chemical substructure calculated across a series of public and proprietary mutagenicity databases. This information was pooled across all sources to identify structural classes that activate or deactivate aromatic amine mutagenicity. This structure activity knowledge, in combination with newly released primary aromatic amine data, was incorporated into Leadscope's expert rule-based and statistical-based (Q)SAR models where increased predictive performance was demonstrated.


Asunto(s)
Aminas/toxicidad , Minería de Datos/métodos , Bases del Conocimiento , Mutagénesis , Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Aminas/química , Aminas/clasificación , Animales , Simulación por Computador , Bases de Datos Factuales , Humanos , Modelos Moleculares , Estructura Molecular , Mutágenos/química , Mutágenos/clasificación , Reconocimiento de Normas Patrones Automatizadas , Relación Estructura-Actividad Cuantitativa , Medición de Riesgo
18.
Regul Toxicol Pharmacol ; 73(1): 367-77, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26248005

RESUMEN

The ICH M7 guidelines for the assessment and control of DNA reactive (mutagenic) impurities in pharmaceuticals allows for the consideration of in silico predictions in place of in vitro studies. This represents a significant advance in the acceptance of (Q)SAR models and has resulted from positive interactions between modellers, regulatory agencies and industry with a shared purpose of developing effective processes to minimise risk. This paper discusses key scientific principles that should be applied when evaluating in silico predictions with a focus on accuracy and scientific rigour that will support a consistent and practical route to regulatory submission.


Asunto(s)
Pruebas de Mutagenicidad/métodos , Pruebas de Mutagenicidad/normas , Simulación por Computador/normas , ADN/química , Contaminación de Medicamentos/prevención & control , Mutágenos , Relación Estructura-Actividad Cuantitativa
19.
J Am Assoc Lab Anim Sci ; 54(2): 163-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25836962

RESUMEN

Cancer risk assessment of new pharmaceuticals is crucial to protect public health. However, clinical trials lack the duration needed to clearly detect drug-related tumor emergence, and biomarkers suggestive of increased cancer risk from a drug typically are not measured in clinical trials. Therefore, the carcinogenic potential of a new pharmaceutical is extrapolated predominately based on 2-y bioassays in rats and mice. A key drawback to this practice is that the results are frequently positive for tumors and can be irrelevant to human cancer risk for reasons such as dose, mode of action, and species specificity. Alternative approaches typically strive to reduce, refine, and replace rodents in carcinogenicity assessments by leveraging findings in short-term studies, both in silico and in vivo, to predict the likely tumor outcome in rodents or, more broadly, to identify a cancer risk to patients. Given the complexities of carcinogenesis and the perceived impracticality of assessing risk in the course of clinical trials, studies conducted in animals will likely remain the standard by which potential cancer risks are characterized for new pharmaceuticals in the immediate foreseeable future. However, a weight-of-evidence evaluation based on short-term toxicologic, in silico, and pharmacologic data is a promising approach to identify with reasonable certainty those pharmaceuticals that present a likely cancer risk in humans and, conversely, those that do not present a human cancer risk.


Asunto(s)
Experimentación Animal , Animales de Laboratorio , Pruebas de Carcinogenicidad , Bienestar del Animal , Animales , Bioensayo , Carcinógenos/toxicidad , Ratones , Ratas , Especificidad de la Especie
20.
Bioorg Med Chem ; 21(17): 4923-7, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23896610

RESUMEN

A series of ring-constrained phenylpropyloxyethylamines, partial opioid structure analogs and derivatives of a previously studied sigma (σ) receptor ligand, was synthesized and evaluated at σ and opioid receptors for receptor selectivity. The results of this study identified several compounds with nanomolar affinity at both σ receptor subtypes. Compounds 6 and 9 had the highest selectivity for both σ receptor subtypes, compared to µ opioid receptors. In addition, compounds 6 and 9 significantly reduced the convulsive effects of cocaine in mice, which would be consistent with antagonism of σ receptors.


Asunto(s)
Ciclohexanoles/química , Etilaminas/química , Fenetilaminas/química , Propilaminas/química , Receptores sigma/antagonistas & inhibidores , Animales , Cocaína/química , Cocaína/toxicidad , Convulsivantes/química , Convulsivantes/metabolismo , Convulsivantes/uso terapéutico , Ciclohexanoles/metabolismo , Ciclohexanoles/uso terapéutico , Etilaminas/metabolismo , Etilaminas/uso terapéutico , Ratones , Fenetilaminas/metabolismo , Fenetilaminas/uso terapéutico , Propilaminas/metabolismo , Propilaminas/uso terapéutico , Unión Proteica , Receptores sigma/metabolismo , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...