Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Drug Metab Pharmacokinet ; 35(1): 18-29, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31952912

RESUMO

Drug-drug interactions (DDIs) involving drug-metabolizing enzymes and membrane transporters can lead to alteration in substrate drug (victim) exposure, and can influence the pharmacological and toxicological effects. In order to predict DDI potential, it is important to quantitatively characterize the major enzyme(s) and/or transporter(s) involved in the clearance of drugs, in terms of fraction metabolized (fm) and fraction transported (ft). In this review, we discuss a strategy using Extended Clearance Classification System (ECCS) to identify the clearance mechanism(s) early in drug discovery, and subsequently rational staging of in vitro characterization to determine fm and ft. In addition, the examples of complex DDIs due to involvement of transporter-enzyme interplay in the hepatic clearance are discussed.


Assuntos
Enzimas/metabolismo , Fígado/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Taxa de Depuração Metabólica , Preparações Farmacêuticas/metabolismo , Interações Medicamentosas , Humanos
2.
Drug Metab Dispos ; 46(6): 865-878, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29487142

RESUMO

Since the introduction of metabolites in safety testing (MIST) guidance by the Food and Drug Administration in 2008, major changes have occurred in the experimental methods for the identification and quantification of metabolites, ways to evaluate coverage of metabolites, and the timing of critical clinical and nonclinical studies to generate this information. In this cross-industry review, we discuss how the increased focus on human drug metabolites and their potential contribution to safety and drug-drug interactions has influenced the approaches taken by industry for the identification and quantitation of human drug metabolites. Before the MIST guidance was issued, the method of choice for generating comprehensive metabolite profile was radio chromatography. The MIST guidance increased the focus on human drug metabolites and their potential contribution to safety and drug-drug interactions and led to changes in the practices of drug metabolism scientists. In addition, the guidance suggested that human metabolism studies should also be accelerated, which has led to more frequent determination of human metabolite profiles from multiple ascending-dose clinical studies. Generating a comprehensive and quantitative profile of human metabolites has become a more urgent task. Together with technological advances, these events have led to a general shift of focus toward earlier human metabolism studies using high-resolution mass spectrometry and to a reduction in animal radiolabel absorption/distribution/metabolism/excretion studies. The changes induced by the MIST guidance are highlighted by six case studies included herein, reflecting different stages of implementation of the MIST guidance within the pharmaceutical industry.


Assuntos
Descoberta de Drogas/normas , Inativação Metabólica/fisiologia , Preparações Farmacêuticas/metabolismo , Animais , Indústria Farmacêutica/normas , Interações Medicamentosas/fisiologia , Humanos , Estados Unidos , United States Food and Drug Administration
3.
Bioanalysis ; 6(5): 641-50, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24620806

RESUMO

We previously developed an high-performance LC-MS peak area ratio approach to demonstrate whether an animal species used in a toxicology study has greater exposures to drug metabolites relative to humans, meeting regulatory guidances regarding safety assessment of drug metabolites. Herein we explain the underlying bioanalytical principals, how to establish all fundamental bioanalytical parameters, and how to evaluate data quality in sample analysis, in the absence of authentic standards of analyte(s). A data-driven tiered approach was used in which data from the peak area ratio method can stand based on statistical analysis, as well as assuring that fundamental elements of bioanalytical method and bioanalysis are met. This strategy offers considerable time- and resource-saving advantage while providing high confidence in the safety assessment of human metabolites in drug development.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Espectrometria de Massas em Tandem/métodos , Animais , Desenho de Fármacos , Humanos
4.
AAPS J ; 15(4): 970-3, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23821354

RESUMO

In the 2012 AAPS metabolites in safety testing (MIST) symposium held in Chicago, IL, USA, on October 15, 2012, regulatory experts and industrial scientists joined together to discuss their perspectives and strategies in addressing contemporary MIST recommendations (FDA 2008, International Conference on Harmonization (ICH) M3(R2), ICH M(R2) Q&A). Overall, these regulatory guidances indicate that metabolites identified in human plasma should circulate at similar or greater concentrations in at least one of the animal species used in nonclinical safety assessment of the parent drug. However, synthetic standards for the metabolites often do not exist or they are intractable to synthesize, thus introducing multiple challenges in drug development for the quantitative comparison of metabolites between human and animals. A tiered bioanalytical strategy for metabolite analysis is a prevalent approach to demonstrate coverage in animals. Recent developments in bioanalytical methodology have yielded several time- and resource-sparing strategies to provide fit-for-purpose approaches that can enable critical decisions related to metabolite quantification and monitoring in plasma. This report summarizes the presentations and panel discussions at the symposium.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/metabolismo , Preparações Farmacêuticas/metabolismo , Animais , Chicago , Avaliação Pré-Clínica de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos/normas , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/sangue , Humanos , Preparações Farmacêuticas/sangue , Especificidade da Espécie
5.
J Clin Pharmacol ; 53(2): 167-77, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23436262

RESUMO

The authors present a comprehensive analysis on the estimation of volume of distribution at steady state (VD(ss) ) in human based on rat, dog, and monkey data on nearly 400 compounds for which there are also associated human data. This data set, to the authors- knowledge, is the largest publicly available, has been carefully compiled from literature reports, and was expanded with some in-house determinations such as plasma protein binding data. This work offers a good statistical basis for the evaluation of applicable prediction methods, their accuracy, and some methods-dependent diagnostic tools. The authors also grouped the compounds according to their charge classes and show the applicability of each method considered to each class, offering further insight into the probability of a successful prediction. Furthermore, they found that the use of fraction unbound in plasma, to obtain unbound volume of distribution, is generally detrimental to accuracy of several methods, and they discuss possible reasons. Overall, the approach using dog and monkey data in the íie-Tozer equation offers the highest probability of success, with an intrinsic diagnostic tool based on aberrant values (<0 or >1) for the calculated fraction unbound in tissue. Alternatively, methods based on dog data (single-species scaling) and rat and dog data (íie-Tozer equation with 2 species or multiple regression methods) may be considered reasonable approaches while not requiring data in nonhuman primates.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Farmacocinética , Animais , Cães , Haplorrinos , Humanos , Ratos , Especificidade da Espécie , Distribuição Tecidual
6.
J Clin Pharmacol ; 53(2): 178-91, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23436263

RESUMO

A comprehensive analysis on the prediction of human clearance based on intravenous pharmacokinetic data from rat, dog, and monkey for approximately 400 compounds was undertaken. This data set has been carefully compiled from literature reports and expanded with some in-house determinations for plasma protein binding and rat clearance. To the authors- knowledge, this is the largest publicly available data set. The present examination offers a comparison of 37 different methods for prediction of human clearance across compounds of diverse physicochemical properties. Furthermore, this work demonstrates the application of each prediction method to each charge class of the compounds, thus presenting an additional dimension to prediction of human pharmacokinetics. In general, the observations suggest that methods employing monkey clearance values and a method incorporating differences in plasma protein binding between rat and human yield the best overall predictions as suggested by approximately 60% compounds within 2-fold geometric mean-fold error. Other single-species scaling or proportionality methods incorporating the fraction unbound in the corresponding preclinical species for prediction of free clearance in human were generally unsuccessful.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Farmacocinética , Animais , Cães , Haplorrinos , Humanos , Taxa de Depuração Metabólica , Ratos , Especificidade da Espécie
7.
Nat Rev Drug Discov ; 10(4): 292-306, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21455238

RESUMO

The normal metabolism of drugs can generate metabolites that have intrinsic chemical reactivity towards cellular molecules, and therefore have the potential to alter biological function and initiate serious adverse drug reactions. Here, we present an assessment of the current approaches used for the evaluation of chemically reactive metabolites. We also describe how these approaches are being used within the pharmaceutical industry to assess and minimize the potential of drug candidates to cause toxicity. At early stages of drug discovery, iteration between medicinal chemistry and drug metabolism can eliminate perceived reactive metabolite-mediated chemical liabilities without compromising pharmacological activity or the need for extensive safety evaluation beyond standard practices. In the future, reactive metabolite evaluation may also be useful during clinical development for improving clinical risk assessment and risk management. Currently, there remains a huge gap in our understanding of the basic mechanisms that underlie chemical stress-mediated adverse reactions in humans. This review summarizes our views on this complex topic, and includes insights into practices considered by the pharmaceutical industry.


Assuntos
Desenho de Fármacos , Descoberta de Drogas/métodos , Preparações Farmacêuticas/metabolismo , Animais , Indústria Farmacêutica/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Medição de Risco/métodos , Gestão de Riscos/métodos
8.
Xenobiotica ; 41(8): 605-22, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21446837

RESUMO

The article describes and discusses the evolution of strategies to characterize metabolites in support of safety studies over the last 40 years, as well as future trends. Approaches to derive qualitative and quantitative information on metabolites are described, with a particular focus on the comparison of options to quantify metabolites in the absence of authentic standards. Current strategies to assess metabolite profiles are summarized into four general approaches and compared against a number of key criteria. Potential future strategies are discussed, including the use of clinical samples as the starting point for metabolite investigations, minimizing the need for animal radiolabelled studies and establishing metabolite safety without radiolabelled studies in animals or human.


Assuntos
Indústria Farmacêutica/tendências , Preparações Farmacêuticas/metabolismo , Animais , Humanos , Espectrometria de Massas , Nitrilas/química , Nitrilas/metabolismo , Pirazóis/química , Pirazóis/metabolismo
9.
Drug Metab Dispos ; 38(10): 1900-5, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20595376

RESUMO

Metabolism is one of the important determinants of the overall disposition of drugs, and the profile of metabolites can have an impact on efficacy and safety. Predicting which drug metabolites will be quantitatively predominant in humans has become increasingly important in the research and development of new drugs. In this study, a novel micropatterned hepatocyte coculture system was evaluated for its ability to generate human in vivo metabolites. Twenty-seven compounds of diverse chemical structure and subject to a range of drug biotransformation reactions were assessed for metabolite profiles in the micropatterned coculture system using pooled cryopreserved human hepatocytes. The ability of this system to generate metabolites that are >10% of dose in excreta or >10% of total drug-related material in circulation was assessed and compared to previously reported data obtained in human hepatocyte suspensions, liver S-9 fraction, and liver microsomes. The micropatterned coculture system was incubated for up to 7 days without a change in medium, which offered an ability to generate metabolites for slowly metabolized compounds. The micropatterned coculture system generated 82% of the excretory metabolites that exceed 10% of dose and 75% of the circulating metabolites that exceed 10% of total circulating drug-related material, exceeds the performance of hepatocyte suspension incubations and other in vitro systems. Phase 1 and phase 2 metabolites were generated, as well as metabolites that arise via two or more sequential reactions. These results suggest that this in vitro system offers the highest performance among in vitro metabolism systems to predict major human in vivo metabolites.


Assuntos
Técnicas de Cocultura/métodos , Hepatócitos/metabolismo , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Criopreservação , Humanos , Desintoxicação Metabólica Fase I , Desintoxicação Metabólica Fase II , Estrutura Molecular , Preparações Farmacêuticas/química , Valor Preditivo dos Testes , Espectrometria de Massas em Tandem
10.
Drug Metab Dispos ; 37(7): 1355-70, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19359406

RESUMO

Time-dependent inhibition (TDI) of cytochrome P450 (P450) enzymes caused by new molecular entities (NMEs) is of concern because such compounds can be responsible for clinically relevant drug-drug interactions (DDI). Although the biochemistry underlying mechanism-based inactivation (MBI) of P450 enzymes has been generally understood for several years, significant advances have been made only in the past few years regarding how in vitro time-dependent inhibition data can be used to understand and predict clinical DDI. In this article, a team of scientists from 16 pharmaceutical research organizations that are member companies of the Pharmaceutical Research and Manufacturers of America offer a discussion of the phenomenon of TDI with emphasis on the laboratory methods used in its measurement. Results of an anonymous survey regarding pharmaceutical industry practices and strategies around TDI are reported. Specific topics that still possess a high degree of uncertainty are raised, such as parameter estimates needed to make predictions of DDI magnitude from in vitro inactivation parameters. A description of follow-up mechanistic experiments that can be done to characterize TDI are described. A consensus recommendation regarding common practices to address TDI is included, the salient points of which include the use of a tiered approach wherein abbreviated assays are first used to determine whether NMEs demonstrate TDI or not, followed by more thorough inactivation studies for those that do to define the parameters needed for prediction of DDI.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Indústria Farmacêutica , Interações Medicamentosas , Microssomos Hepáticos/metabolismo , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP3A/metabolismo , Desenho de Fármacos , Glucuronosiltransferase , Humanos , Microssomos Hepáticos/enzimologia , Oxirredutases N-Desmetilantes/metabolismo , Preparações Farmacêuticas/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Fatores de Tempo
11.
Chem Res Toxicol ; 22(2): 348-56, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19170655

RESUMO

The role of metabolism in genotoxicity and carcinogenicity of many chemicals is well established. Accordingly, both in vitro metabolic activation systems and in vivo assays are routinely utilized for genotoxic hazard identification of drug candidates prior to clinical investigations. This should, in most cases provide a high degree of confidence that the genotoxic potential of the parent and associated metabolites have been characterized. However, it is well known that significant differences can exist between human metabolism and that which occurs with in vitro and in vivo genotoxicity tests. This poses challenges when considering the adequacy of hazard identification and cancer risk assessment if a human metabolite of genotoxic concern is identified during the course of drug development. Since such challenges are particularly problematic when recognized in the later stages of drug development, a framework for conducting a carcinogenic risk assessment for human genotoxic metabolites is desirable. Here, we propose a risk assessment method that is dependent upon the availability of quantitative human and rodent ADME (absorption, distribution, metabolism, excretion) data, such that exposures to a metabolite of genotoxic concern can be estimated at the intended human efficacious dose and the maximum dose used in the 2-year rodent bioassay(s). The exposures are then applied to the risk assessment framework, based on known cancer potencies, that allows one to understand the probability of a known or suspect genotoxic metabolite posing a carcinogenic risk in excess of 1 in 100,000. Practical case examples are presented to illustrate the application of the risk assessment method within the context of drug development and to highlight its utility and limitations.


Assuntos
Testes de Mutagenicidade/métodos , Mutagênicos/metabolismo , Mutagênicos/toxicidade , Animais , Carga Corporal (Radioterapia) , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Humanos , Redes e Vias Metabólicas , Ratos , Medição de Risco
12.
Chem Res Toxicol ; 22(2): 357-68, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19146377

RESUMO

An early understanding of key metabolites of drugs is crucial in drug discovery and development. As a result, several in vitro models typically derived from liver are frequently used to study drug metabolism. It is presumed that these in vitro systems provide an accurate view of the potential in vivo metabolites and metabolic pathways. However, no formal analysis has been conducted to validate their use. The goal of the present study was to conduct a comprehensive analysis to assess if the three commonly used in vitro systems, pooled human liver microsomes, liver S-9 fraction, and hepatocytes, adequately predict in vivo metabolic profiles for drugs. The second objective was to compare the overall capabilities of these three systems to generate in vivo metabolic profiles. Twenty-seven compounds in the Pfizer database and 21 additional commercially available compounds of diverse structure and routes of metabolism for which the human ADME data was available were analyzed in this study to assess the performance of the in vitro systems. The results suggested that all three systems reliably predicted human excretory and circulating metabolite profiles. Furthermore, the success in predicting primary metabolites and metabolic pathways was high (>70%), but the predictability of secondary metabolites was less reliable in the three systems. Thus, the analysis provides sufficient confidence in using in vitro systems to reliably produce primary in vivo human metabolites and supports their application in early discovery to identify metabolic spots for optimization of metabolic liabilities anticipated in humans in vivo. However, the in vitro systems cannot solely mitigate the risk of disproportionate circulating metabolites in humans and may need to be supplemented with metabolic profiling of plasma samples from first-in-human studies or early human radiolabeled studies.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Hepatócitos/metabolismo , Redes e Vias Metabólicas , Microssomos Hepáticos/metabolismo , Preparações Farmacêuticas/metabolismo , Descoberta de Drogas , Indústria Farmacêutica , Humanos , Preparações Farmacêuticas/sangue , Preparações Farmacêuticas/urina
13.
Environ Mol Mutagen ; 49(8): 631-41, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18626997

RESUMO

Because it is well known that metabolites of chemicals and drugs are frequently the ultimate species responsible for genotoxicity and carcinogenicity, in vitro testing to identify the human genotoxicity hazard potential of new chemicals and drugs routinely utilizes liver S-9 fraction from rats treated with Aroclor 1254 as a system that can generate metabolites. However, it is frequently questioned as to whether such an in vitro metabolite generation system is the most relevant for human risk, or whether the assay would be better served by using a human-derived in vitro system. To address this, 16 common drugs have been examined for profiles of metabolites in Aroclor-induced rat liver S-9 and pooled human liver S-9. Metabolite profiles were compared using high pressure liquid chromatography coupled with ion trap mass spectrometry, in line with ultraviolet or radiometric detection to help make semiquantitative comparisons. Results showed that, with few exceptions, metabolites generated in the human system were also generated in the rat system. Also, in several cases the rat system generated considerably more metabolites, suggesting that there is a potential that positive genotoxicity findings could be caused by metabolites that have no relevance to humans. These findings suggest that when conducting in vitro genotoxicity testing using the Aroclor-induced rat liver S-9 system, knowledge of the metabolite profile in the system is important, and a comparison to the profile generated in human liver S-9 could be of value when interpreting the genotoxicity results.


Assuntos
Arocloros/toxicidade , Fígado/efeitos dos fármacos , Mutagênicos/toxicidade , Frações Subcelulares/efeitos dos fármacos , Animais , Arocloros/farmacocinética , Biotransformação , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Fígado/enzimologia , Fígado/ultraestrutura , Testes de Mutagenicidade , Ratos
14.
Curr Drug Metab ; 8(5): 449-62, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17584016

RESUMO

This commentary discusses the approaches to, and key considerations in the in vitro-in vivo extrapolation of drug-drug interactions (DDI) resulting from mechanism-based inactivation (MBI) of cytochrome P450 (CYP) enzymes and clinical pharmacologic implications. In vitro kinetic assessment and prediction of DDI produced via reversible inhibition and MBI rely on operationally and conceptually distinct approaches. DDI risk assessment for inactivators requires estimation of maximal inactivation rate (k(inact)) and inactivator potency (KI) in vitro, that need to be considered in context of the biological turnover rate of the enzyme (kdeg) and clinical exposures of the inactivator (I), respectively, to predict interaction magnitude. Risk assessment cannot be performed by a simple comparison of inactivator potency against in vivo exposure since inactivation is both concentration and time-dependent. MBI contour plots tracking combinations of I:KI and k(inact):k(deg) resulting in identical fold-reductions in intrinsic clearance are proposed as a useful framework for DDI risk assessment. Additionally, substrate-specific factors like fraction of the total clearance of the object drug via the enzyme being inactivated (f(m(CYP) )) and the bioavailability fraction across the intestine for CYP3A substrates (F(G)) are important determinants of interaction magnitude. Sensitivity analysis of predicted DDI magnitude to uncertainty in input parameters is recommended to inform confidence in predictions. The time course of reversal of DDI resulting from CYP inactivation is determined by the half-life of the enzyme which is an important consideration in the design and interpretation of clinical DDI studies with inactivators.


Assuntos
Inibidores das Enzimas do Citocromo P-450 , Xenobióticos/efeitos adversos , Xenobióticos/farmacologia , Interações Medicamentosas , Humanos , Concentração Inibidora 50 , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Modelos Biológicos , Valor Preditivo dos Testes , Medição de Risco , Especificidade por Substrato
15.
Mutat Res ; 627(1): 59-77, 2007 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-17141553

RESUMO

The report from the 2002 International Workshop on Genotoxicity Tests (IWGT) Strategy Expert Group emphasized metabolic considerations as an important area to address in developing a common strategy for genotoxicity testing. A working group convened at the 2005 4th IWGT to discuss this area further and propose practical strategy recommendations. To propose a strategy, the working group reviewed: (1) the current status and deficiencies, including examples of carcinogens "missed" in genotoxicity testing, established shortcomings of the standard in vitro induced S9 activation system and drug metabolite case examples; (2) the current status of possible remedies, including alternative S9 sources, other external metabolism systems or genetically engineered test systems; (3) any existing positions or guidance. The working group established consensus principles to guide strategy development. Thus, a human metabolite of interest should be represented in genotoxicity and carcinogenicity testing, including evaluation of alternative genotoxicity in vitro metabolic activation or test systems, and the selection of a carcinogenicity test species showing appropriate biotransformation. Appropriate action triggers need to be defined based on the extent of human exposure, considering any structural knowledge of the metabolite, and when genotoxicity is observed upon in vitro testing in the presence of metabolic activation. These triggers also need to be considered in defining the timing of human pharmaceutical ADME assessments. The working group proposed two strategies to consider; a more proactive approach, which emphasizes early metabolism predictions to drive appropriate hazard assessment; and a retroactive approach to manage safety risks of a unique or "major" metabolite once identified and quantitated from human clinical ADME studies. In both strategies, the assessment of the genotoxic potential of a metabolite could include the use of an alternative or optimized in vitro metabolic activation system, or direct testing of an isolated or synthesized metabolite. The working group also identified specific areas where more data or experiences need to be gained to reach consensus. These included defining a discrete exposure action trigger for safety assessment and when direct testing of a metabolite of interest is warranted versus the use of an alternative in vitro activation system, a universal recommendation for the timing of human ADME studies for drug candidates and the positioning of metabolite structural knowledge (through in silico systems, literature, expert analysis) in supporting metabolite safety qualification. Lastly, the working group outlined future considerations for refining the initially proposed strategies. These included the need for further evaluation of the current in vitro genotoxicity testing protocols that can potentially perturb or reduce the level of metabolic activity (potential alterations in metabolism associated with both the use of some solvents to solubilize test chemicals and testing to the guidance limit dose), and proposing broader evaluations of alternative metabolic activation sources or engineered test systems to further challenge the suitability of (or replace) the current induced liver S9 activation source.


Assuntos
Redes e Vias Metabólicas , Testes de Mutagenicidade/métodos , 2-Acetilaminofluoreno/metabolismo , 2-Acetilaminofluoreno/toxicidade , Animais , Carcinógenos/toxicidade , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Indústria Farmacêutica , Enzimas/química , Guias como Assunto , Humanos , Fígado/metabolismo , Testes de Mutagenicidade/normas , Testes de Mutagenicidade/tendências , Extratos Vegetais/metabolismo , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solventes/química , Estados Unidos , United States Food and Drug Administration
16.
Curr Drug Metab ; 4(6): 527-34, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14683480

RESUMO

For the pharmaceutical industry, one of the challenges in evaluating the risk of future compound attrition at the discovery stage is the successful prediction of the major routes of clearance in humans. For compounds cleared by metabolism, such information will help to avoid the development of compounds that will exhibit large interpatient differences in pharmacokinetics via 1). routes of metabolism catalyzed by functionally polymorphic enzymes and/or 2). clinically significant metabolic drug-drug interactions, in the later stages of development. The degree of intersubject variability that is acceptable for a drug candidate is uncertain in the discovery stage where knowledge of other important factors is limited or unavailable (i.e. therapeutic index, pharmacodynamic variability, etc). Reaction phenotyping is the semi-quantitative in vitro estimation of the relative contributions of specific drug-metabolizing enzymes to the metabolism of a test compound. However, reaction phenotyping in the discovery stage of drug development is complicated by the absence of radiolabelled parent compound or metabolite bioanalytical standards relative to later stages of development. In this commentary, some of the approaches, based on published data, which can be taken to overcome these challenges are discussed. In addition, knowledge of the molecular structure (i.e. specific chemical substituents), physicochemical properties, and routes of clearance in animals can all help in making a successful prediction for the routes of clearance in humans. In combination, the objective of these studies should be to reduce to a minimum the risk of finding significant inter-patient differences in pharmacokinetics at a later stage in development due to significant metabolism by polymorphic enzymes or drug-drug interactions. Consequently, this data should be used to avoid costly late stage attrition.


Assuntos
Desenho de Fármacos , Fenótipo , Sistema Enzimático do Citocromo P-450/metabolismo , Indústria Farmacêutica , Humanos , Preparações Farmacêuticas/metabolismo
17.
Drug Metab Dispos ; 31(7): 815-32, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12814957

RESUMO

Current regulatory guidances do not address specific study designs for in vitro and in vivo drug-drug interaction studies. There is a common desire by regulatory authorities and by industry sponsors to harmonize approaches, to allow for a better assessment of the significance of findings across different studies and drugs. There is also a growing consensus for the standardization of cytochrome P450 (P450) probe substrates, inhibitors and inducers and for the development of classification systems to improve the communication of risk to health care providers and to patients. While existing guidances cover mainly P450-mediated drug interactions, the importance of other mechanisms, such as transporters, has been recognized more recently, and should also be addressed. This article was prepared by the Pharmaceutical Research and Manufacturers of America (PhRMA) Drug Metabolism and Clinical Pharmacology Technical Working Groups and represents the current industry position. The intent is to define a minimal best practice for in vitro and in vivo pharmacokinetic drug-drug interaction studies targeted to development (not discovery support) and to define a data package that can be expected by regulatory agencies in compound registration dossiers.


Assuntos
Indústria Farmacêutica , Interações Medicamentosas , Projetos de Pesquisa , Sistema Enzimático do Citocromo P-450/classificação , Sistema Enzimático do Citocromo P-450/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA