Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Regul Toxicol Pharmacol ; 150: 105640, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38754805

RESUMEN

N-Nitrosamine impurities, including nitrosamine drug substance-related impurities (NDSRIs), have challenged pharmaceutical industry and regulators alike and affected the global drug supply over the past 5 years. Nitrosamines are a class of known carcinogens, but NDSRIs have posed additional challenges as many lack empirical data to establish acceptable intake (AI) limits. Read-across analysis from surrogates has been used to identify AI limits in some cases; however, this approach is limited by the availability of robustly-tested surrogates matching the structural features of NDSRIs, which usually contain a diverse array of functional groups. Furthermore, the absence of a surrogate has resulted in conservative AI limits in some cases, posing practical challenges for impurity control. Therefore, a new framework for determining recommended AI limits was urgently needed. Here, the Carcinogenic Potency Categorization Approach (CPCA) and its supporting scientific rationale are presented. The CPCA is a rapidly-applied structure-activity relationship-based method that assigns a nitrosamine to 1 of 5 categories, each with a corresponding AI limit, reflecting predicted carcinogenic potency. The CPCA considers the number and distribution of α-hydrogens at the N-nitroso center and other activating and deactivating structural features of a nitrosamine that affect the α-hydroxylation metabolic activation pathway of carcinogenesis. The CPCA has been adopted internationally by several drug regulatory authorities as a simplified approach and a starting point to determine recommended AI limits for nitrosamines without the need for compound-specific empirical data.


Asunto(s)
Carcinógenos , Contaminación de Medicamentos , Nitrosaminas , Nitrosaminas/análisis , Nitrosaminas/toxicidad , Carcinógenos/análisis , Carcinógenos/toxicidad , Contaminación de Medicamentos/prevención & control , Humanos , Animales , Relación Estructura-Actividad , Medición de Riesgo , Pruebas de Carcinogenicidad
2.
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.

3.
Regul Toxicol Pharmacol ; 125: 105006, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34273441

RESUMEN

The ICH M7 (R1) guideline recommends the use of complementary (Q)SAR models to assess the mutagenic potential of drug impurities as a state-of-the-art, high-throughput alternative to empirical testing. Additionally, it includes a provision for the application of expert knowledge to increase prediction confidence and resolve conflicting calls. Expert knowledge, which considers structural analogs and mechanisms of activity, has been valuable when models return an indeterminate (equivocal) result or no prediction (out-of-domain). A retrospective analysis of 1002 impurities evaluated in drug regulatory applications between April 2017 and March 2019 assessed the impact of expert review on (Q)SAR predictions. Expert knowledge overturned the default predictions for 26% of the impurities and resolved 91% of equivocal predictions and 75% of out-of-domain calls. Of the 261 overturned default predictions, 15% were upgraded to equivocal or positive and 79% were downgraded to equivocal or negative. Chemical classes with the most overturns were primary aromatic amines (46%), aldehydes (45%), Michael-reactive acceptors (37%), and non-primary alkyl halides (33%). Additionally, low confidence predictions were the most often overturned. Collectively, the results suggest that expert knowledge continues to play an important role in an ICH M7 (Q)SAR prediction workflow and triaging predictions based on chemical class and probability can improve (Q)SAR review efficiency.


Asunto(s)
Contaminación de Medicamentos , Mutágenos/química , Relación Estructura-Actividad Cuantitativa , Simulación por Computador , Pruebas de Mutagenicidad , Estudios Retrospectivos , Medición de Riesgo
4.
Comput Toxicol ; 202021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35368437

RESUMEN

Historically, identifying carcinogens has relied primarily on tumor studies in rodents, which require enormous resources in both money and time. In silico models have been developed for predicting rodent carcinogens but have not yet found general regulatory acceptance, in part due to the lack of a generally accepted protocol for performing such an assessment as well as limitations in predictive performance and scope. There remains a need for additional, improved in silico carcinogenicity models, especially ones that are more human-relevant, for use in research and regulatory decision-making. As part of an international effort to develop in silico toxicological protocols, a consortium of toxicologists, computational scientists, and regulatory scientists across several industries and governmental agencies evaluated the extent to which in silico models exist for each of the recently defined 10 key characteristics (KCs) of carcinogens. This position paper summarizes the current status of in silico tools for the assessment of each KC and identifies the data gaps that need to be addressed before a comprehensive in silico carcinogenicity protocol can be developed for regulatory use.

5.
Regul Toxicol Pharmacol ; 118: 104807, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33058939

RESUMEN

Pharmaceutical applicants conduct (Q)SAR assessments on identified and theoretical impurities to predict their mutagenic potential. Two complementary models-one rule-based and one statistical-based-are used, followed by expert review. (Q)SAR models are continuously updated to improve predictions, with new versions typically released on a yearly basis. Numerous releases of (Q)SAR models will occur during the typical 6-7 years of drug development until new drug registration. Therefore, it is important to understand the impact of model updates on impurity mutagenicity predictions over time. Compounds representative of pharmaceutical impurities were analyzed with three rule- and three statistical-based models covering a 4-8 year period, with the individual time frame being dependent on when the individual models were initially made available. The largest changes in the combined outcome of two complementary models were from positive or equivocal to negative and from negative to equivocal. Importantly, the cumulative change of negative to positive predictions was small in all models (<5%) and was further reduced when complementary models were combined in a consensus fashion. We conclude that model updates of the type evaluated in this manuscript would not necessarily require re-running a (Q)SAR prediction unless there is a specific need. However, original (Q)SAR predictions should be evaluated when finalizing the commercial route of synthesis for marketing authorization.


Asunto(s)
Contaminación de Medicamentos , Desarrollo de Medicamentos , Modelos Moleculares , Pruebas de Mutagenicidad , Preparaciones Farmacéuticas/análisis , Programas Informáticos , Animales , Simulación por Computador , Humanos , Relación Estructura-Actividad Cuantitativa , Medición de Riesgo , Factores de Tiempo , Flujo de Trabajo
6.
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
7.
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
8.
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
9.
Regul Toxicol Pharmacol ; 107: 104403, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31195068

RESUMEN

In silico toxicology (IST) approaches to rapidly assess chemical hazard, and usage of such methods is increasing in all applications but especially for regulatory submissions, such as for assessing chemicals under REACH as well as the ICH M7 guideline for drug impurities. There are a number of obstacles to performing an IST assessment, including uncertainty in how such an assessment and associated expert review should be performed or what is fit for purpose, as well as a lack of confidence that the results will be accepted by colleagues, collaborators and regulatory authorities. To address this, a project to develop a series of IST protocols for different hazard endpoints has been initiated and this paper describes the genetic toxicity in silico (GIST) protocol. The protocol outlines a hazard assessment framework including key effects/mechanisms and their relationships to endpoints such as gene mutation and clastogenicity. IST models and data are reviewed that support the assessment of these effects/mechanisms along with defined approaches for combining the information and evaluating the confidence in the assessment. This protocol has been developed through a consortium of toxicologists, computational scientists, and regulatory scientists across several industries to support the implementation and acceptance of in silico approaches.


Asunto(s)
Modelos Teóricos , Mutágenos/toxicidad , Proyectos de Investigación , Toxicología/métodos , Animales , Simulación por Computador , Humanos , Pruebas de Mutagenicidad , Medición de Riesgo
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Regul Toxicol Pharmacol ; 96: 1-17, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29678766

RESUMEN

The present publication surveys several applications of in silico (i.e., computational) toxicology approaches across different industries and institutions. It highlights the need to develop standardized protocols when conducting toxicity-related predictions. This contribution articulates the information needed for protocols to support in silico predictions for major toxicological endpoints of concern (e.g., genetic toxicity, carcinogenicity, acute toxicity, reproductive toxicity, developmental toxicity) across several industries and regulatory bodies. Such novel in silico toxicology (IST) protocols, when fully developed and implemented, will ensure in silico toxicological assessments are performed and evaluated in a consistent, reproducible, and well-documented manner across industries and regulatory bodies to support wider uptake and acceptance of the approaches. The development of IST protocols is an initiative developed through a collaboration among an international consortium to reflect the state-of-the-art in in silico toxicology for hazard identification and characterization. A general outline for describing the development of such protocols is included and it is based on in silico predictions and/or available experimental data for a defined series of relevant toxicological effects or mechanisms. The publication presents a novel approach for determining the reliability of in silico predictions alongside experimental data. In addition, we discuss how to determine the level of confidence in the assessment based on the relevance and reliability of the information.


Asunto(s)
Simulación por Computador , Pruebas de Toxicidad/métodos , Toxicología/métodos , Animales , Humanos
16.
Ther Innov Regul Sci ; 52(2): 244-255, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29568713

RESUMEN

In 2011, the US Food and drug Administration (FDA) developed a strategic plan for regulatory science that focuses on developing new tools, standards, and approaches to assess the safety, efficacy, quality, and performance of FDA-regulated products. In line with this, the Division of Applied Regulatory Science was created to move new science into the Center for Drug Evaluation and Research (CDER) review process and close the gap between scientific innovation and drug review. The Division, located in the Office of Clinical Pharmacology, is unique in that it performs mission-critical applied research and review across the translational research spectrum including in vitro and in vivo laboratory research, in silico computational modeling and informatics, and integrated clinical research covering clinical pharmacology, experimental medicine, and postmarket analyses. The Division collaborates with Offices throughout CDER, across the FDA, other government agencies, academia, and industry. The Division is able to rapidly form interdisciplinary teams of pharmacologists, biologists, chemists, computational scientists, and clinicians to respond to challenging regulatory questions for specific review issues and for longer-range projects requiring the development of predictive models, tools, and biomarkers to speed the development and regulatory evaluation of safe and effective drugs. This article reviews the Division's recent work and future directions, highlighting development and validation of biomarkers; novel humanized animal models; translational predictive safety combining in vitro, in silico, and in vivo clinical biomarkers; chemical and biomedical informatics tools for safety predictions; novel approaches to speed the development of complex generic drugs, biosimilars, and antibiotics; and precision medicine.

17.
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
18.
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
19.
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
20.
Int J Toxicol ; 34(4): 352-4, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25979517

RESUMEN

A continuing education (CE) course at the 2014 American College of Toxicology annual meeting covered the topic of (Quantitative) Structure-Activity Relationships [(Q)SAR]. The (Q)SAR methodologies use predictive computer modeling based on predefined rules to describe the relationship between chemical structure and a chemical's associated biological activity or statistical tools to find correlations between biologic activity and the molecular structure or properties of a compound. The (Q)SAR has applications in risk assessment, drug discovery, and regulatory decision making. Pressure within industry to reduce the cost of drug development and societal pressure for government regulatory agencies to produce more accurate and timely risk assessment of drugs and chemicals have necessitated the use of (Q)SAR. Producing a high-quality (Q)SAR model depends on many factors including the choice of statistical methods and descriptors, but first and foremost the quality of the data input into the model. Understanding how a (Q)SAR model is developed and applied is critical to the successful use of such a tool. The CE session covered the basic principles of (Q)SAR, practical applications of these computational models in toxicology, how regulatory agencies use and interpret (Q)SAR models, and potential pitfalls of using them.


Asunto(s)
Relación Estructura-Actividad Cuantitativa , Pruebas de Toxicidad/métodos , Simulación por Computador , Congresos como Asunto , Humanos , Estructura Molecular , Medición de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA