Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Regul Toxicol Pharmacol ; 117: 104746, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32911461

RESUMO

Pharmaceutic products designed to perturb the function of epigenetic modulators have been approved by regulatory authorities for treatment of advanced cancer. While the predominant effort in epigenetic drug development continues to be in oncology, non-oncology indications are also garnering interest. A survey of pharmaceutical companies was conducted to assess the interest and concerns for developing small molecule direct epigenetic effectors (EEs) as medicines. Survey themes addressed (1) general levels of interest and activity with EEs as therapeutic agents, (2) potential safety concerns, and (3) possible future efforts to develop targeted strategies for nonclinical safety assessment of EEs. Thirteen companies contributed data to the survey. Overall, the survey data indicate the consensus opinion that existing ICH guidelines are effective and appropriate for nonclinical safety assessment activities with EEs. Attention in the framework of study design should, on a case by case basis, be considered for delayed or latent toxicities, carcinogenicity, reproductive toxicity, and the theoretical potential for transgenerational effects. While current guidelines have been appropriate for the nonclinical safety assessments of epigenetic targets, broader experience with a wide range of epigenetic targets will provide information to assess the potential need for new or revised risk assessment strategies for EE drugs.


Assuntos
Indústria Farmacêutica/normas , Controle de Medicamentos e Entorpecentes , Epigênese Genética/efeitos dos fármacos , Preparações Farmacêuticas/normas , Inquéritos e Questionários , Animais , Avaliação Pré-Clínica de Medicamentos/normas , Avaliação Pré-Clínica de Medicamentos/tendências , Indústria Farmacêutica/tendências , Controle de Medicamentos e Entorpecentes/tendências , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Epigênese Genética/genética , Humanos , Preparações Farmacêuticas/administração & dosagem , Medição de Risco/normas , Medição de Risco/tendências
2.
Chem Res Toxicol ; 33(1): 125-136, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31840498

RESUMO

Kinase inhibitors have transformed the treatment of many cancers and are showing the same promise for other indications including inflammatory diseases. This class of drugs is one of the most predominant in the pharmaceutical industry, but development and clinical utility is often limited by a broad spectrum of cardiovascular (CV) toxicities including QT prolongation and arrhythmia, left ventricular dysfunction, congestive heart failure, ischemia and myocardial infarction, and hypertension. In this review article, we provide a broad overview of the spectrum of CV events detected in clinical trials of kinase inhibitors and the known and proposed on- and off-target links between kinase inhibitor targets and these specific cardiotoxicities. We also examine the unique features of kinase inhibitors that have impeded complete mechanistic understanding of kinase inhibitor-associated cardiotoxicities and contributed to the disconnect between preclinical predictions and clinical findings. We then discuss various in vitro models currently in use that are amenable for high-throughput screening as well as lower throughput models that are valuable for mechanistic insight. These physiologically relevant models, together with newer "omic"-wide approaches will help to increase our understanding of the mechanisms underlying kinase inhibitor-associated cardiotoxicity and enable rational design of kinase inhibitors in the future.


Assuntos
Cardiotoxicidade/etiologia , Inibidores de Proteínas Quinases/efeitos adversos , Animais , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos
3.
Cardiovasc Toxicol ; 15(2): 127-39, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25134468

RESUMO

Cardiovascular (CV) toxicity is a leading cause of drug attrition and withdrawal. Introducing in vitro assays with higher throughput should permit earlier CV hazard identification and enable medicinal chemists to design-out liabilities. Heretofore, development of in vitro CV assays has been limited by the challenge of replicating integrated cardiovascular physiology while achieving the throughput and consistency required for screening. These challenges appear to be met with a combination of human stem cell-derived cardiomyocytes (CM) which beat spontaneously and monitoring the response with technology that can assess drug-induced changes in voltage dependent contraction such as cellular impedance which has been validated with excellent predictivity for drug-induced arrhythmia and contractility. Here, we review advances in cardiomyocyte impedance with emphasis on stem cell-derived cardiomyocyte models for toxicity screening. Key perspectives include: the electrical principles of impedance technology, impedance detection of cardiomyocyte beating, beat parameter selection/analysis, validation in toxicity and drug discovery, and future directions. As a conclusion, an in vitro screening cascade is proffered using the downstream, inclusive detection of CM impedance assays as a primary screen followed by complementary CM assays chosen to enable mechanism-appropriate follow-up. The combined approach will enhance testing for CV liabilities prior to traditional in vivo models.


Assuntos
Cardiotoxinas/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Cardiotoxicidade/diagnóstico , Cardiotoxicidade/patologia , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Impedância Elétrica , Humanos , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Células-Tronco/patologia , Células-Tronco/fisiologia
4.
Cell Rep ; 9(3): 810-21, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25437537

RESUMO

Diabetic cardiomyopathy is a complication of type 2 diabetes, with known contributions of lifestyle and genetics. We develop environmentally and genetically driven in vitro models of the condition using human-induced-pluripotent-stem-cell-derived cardiomyocytes. First, we mimic diabetic clinical chemistry to induce a phenotypic surrogate of diabetic cardiomyopathy, observing structural and functional disarray. Next, we consider genetic effects by deriving cardiomyocytes from two diabetic patients with variable disease progression. The cardiomyopathic phenotype is recapitulated in the patient-specific cells basally, with a severity dependent on their original clinical status. These models are incorporated into successive levels of a screening platform, identifying drugs that preserve cardiomyocyte phenotype in vitro during diabetic stress. In this work, we present a patient-specific induced pluripotent stem cell (iPSC) model of a complex metabolic condition, showing the power of this technique for discovery and testing of therapeutic strategies for a disease with ever-increasing clinical significance.


Assuntos
Cardiomiopatias Diabéticas/patologia , Avaliação Pré-Clínica de Medicamentos , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Diferenciação Celular/efeitos dos fármacos , Humanos , Hipertrofia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fenótipo , Sarcômeros/efeitos dos fármacos , Sarcômeros/patologia , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
5.
J Am Coll Cardiol ; 61(3): 267-74, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23328609

RESUMO

Cancer genomics has focused on the discovery of mutations and chromosomal structural rearrangements that either increase susceptibility to cancer or support the cancer phenotype. Protein kinases are the most frequently mutated genes in the cancer genome, making them attractive therapeutic targets for drug design. However, the use of some of the kinase inhibitors (KIs) has been associated with toxicities to the heart and vasculature, including acute coronary syndromes and heart failure. Herein we discuss the genetic basis of cancer, focusing on mutations in the kinase genome (kinome) that lead to tumorigenesis. This will allow an understanding of the real and potential power of modern cancer therapeutics. The underlying mechanisms that drive the cardiotoxicity of the KIs are also examined. The preclinical models for predicting cardiotoxicity, including induced pluripotent stem cells and zebrafish, are reviewed, with the hope of eventually being able to identify problematic agents before their use in patients. Finally, the use of biomarkers in the clinic is discussed, and newer strategies (i.e., metabolomics and enhanced imaging strategies) that may allow earlier and more accurate detection of cardiotoxicity are reviewed.


Assuntos
Antineoplásicos/efeitos adversos , Cardiotoxinas/uso terapêutico , Coração/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Inibidores de Proteínas Quinases/efeitos adversos , Animais , Antineoplásicos/farmacologia , Cardiotoxinas/efeitos adversos , Análise Mutacional de DNA , Reparo do DNA , Avaliação Pré-Clínica de Medicamentos/métodos , Genoma Humano , Humanos , Metabolômica , Neoplasias/fisiopatologia , Guias de Prática Clínica como Assunto , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Quinases/efeitos dos fármacos , Transdução de Sinais/genética
7.
Toxicol Sci ; 123(1): 281-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21693436

RESUMO

Improved in vitro systems for predicting drug-induced toxicity are needed in the pharmaceutical and biotechnology industries to decrease late-stage drug attrition. One unmet need is an early screen for cardiotoxicity, which accounts for about one third of safety-based withdrawn pharmaceuticals. Herein, the first published report of a high-throughput functional assay employing a monolayer of beating human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is described, detailing a model that accurately detects drug-induced cardiac abnormalities. Using 96-well plates with interdigitated electrode arrays that assess impedance, the rhythmic, synchronous contractions of the iPSC-CMs were detected. Treatment of the iPSC-CMs with 28 different compounds with known cardiac effects resulted in compound-specific changes in the beat rate and/or the amplitude of the impedance measurement. Changes in impedance for the compounds tested were comparable with the results from a related technology, electric field potential assessment obtained from microelectrode arrays. Using the results from the set of compounds, an index of drug-induced arrhythmias was calculated, enabling the determination of a drug's proarrhythmic potential. This system of interrogating human cardiac function in vitro opens new opportunities for predicting cardiac toxicity and studying cardiac biology.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Avaliação Pré-Clínica de Medicamentos/efeitos adversos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Descoberta de Drogas/métodos , Humanos , Preparações Farmacêuticas
8.
Nat Rev Drug Discov ; 10(2): 111-26, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21283106

RESUMO

Targeted therapeutics, particularly those that inhibit the activity of protein kinases that are mutated and/or overexpressed in cancer, have revolutionized the treatment of some cancers and improved survival rates in many others. Although these agents dominate drug development in cancer, significant toxicities, including cardiotoxicity, have emerged. In this Review, we examine the underlying mechanisms that result in on-target or off-target cardiotoxicities of small molecule kinase inhibitors. We also discuss how well the various preclinical safety models and strategies might predict clinical cardiotoxicity. It is hoped that a thorough understanding of the mechanisms underlying cardiotoxicity will lead to the development of safe, effective drugs and consequently, fewer costly surprises as agents progress through clinical trials.


Assuntos
Cardiotoxinas/uso terapêutico , Ensaios Clínicos como Assunto/métodos , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Cardiotoxinas/efeitos adversos , Cardiotoxinas/farmacocinética , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Valor Preditivo dos Testes , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Resultado do Tratamento
9.
Expert Opin Drug Saf ; 7(2): 107-10, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18324874

RESUMO

The cost impact of late-stage failures of drug candidates has motivated the pharmaceutical industry to develop, validate, and implement a more proactive testing paradigm, including an emphasis on conducting predictive in vitro and in vivo studies earlier. The goal of drug discovery toxicology is not to reduce or eliminate attrition, as is often mis-stated as such, but rather to reprioritize efforts to shift attrition of future failing molecules upstream in discovery. This shift in attrition requires additional studies and investment earlier in the candidate evaluation process in order to avoid spending resources on molecules with soon-to-be-discovered development-limiting liabilities. While in silico and in vitro models will continually be developed and refined, in vivo preclinical safety models remain the gold standard for assessing human risk. For in vivo testing to influence early discovery effectively, it must: i) require low amounts of compound; ii) provide rapid results to drive decision-making and medicinal chemistry efforts; and iii) be flexible and provide results relevant to the development plan tailored to each target, drug class, and/or indication.


Assuntos
Desenho de Fármacos , Drogas em Investigação/efeitos adversos , Drogas em Investigação/toxicidade , Modelos Animais , Testes de Toxicidade , Animais , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos/métodos , Valor Preditivo dos Testes , Especificidade da Espécie , Fatores de Tempo
10.
Mol Pharmacol ; 71(6): 1475-86, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17327465

RESUMO

Expression of Cyp1a1 and its related enzyme activity have long been used as a biomarker for aryl hydrocarbon receptor (AhR) activation and a warning of dioxin-like toxicity. As a result, induction of Cyp1a1 by pharmaceutical drug candidates or environmental contaminants raises significant concern in risk assessment. The current study evaluates the specificity of Cyp1a1 induction as a marker for AhR affinity and activation and provides context to assess the relevancy of AhR activation to risk assessment. In vivo experiments examined the expression of Cyp1a1 and other AhR-regulated genes in liver, kidney, and heart in response to 596 compounds. From this data set, a subset of 147 compounds was then evaluated for their ability to activate or bind to the AhR using a combination of gel shift, reporter gene, and competitive receptor binding assays. Whereas in vivo Cyp1a1 mRNA expression is a sensitive marker for AhR activation, it lacks specificity, because 81 (59%) of 137 compounds were found to significantly induce Cyp1a1 in vivo but were not verified to bind or activate the AhR in vitro. Combining in vivo and in vitro findings, we identified nine AhR agonists, six of which are marketed therapeutics and have been approved by the U.S. Food and Drug Administration, including leflunomide, flutamide, and nimodipine. These drugs do not produce dioxin-like toxicity in rats or in humans. These data demonstrate that induction of Cyp1a1 is a nonspecific biomarker of direct AhR affinity and activation and lend further support to the hypothesis that Cyp1a1 induction and/or AhR activation is not synonymous with dioxin-like toxicity.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Biomarcadores/metabolismo , Citocromo P-450 CYP1A1/genética , Dioxinas/toxicidade , Avaliação Pré-Clínica de Medicamentos , Indução Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Especificidade de Órgãos , Preparações Farmacêuticas , Ratos , Ratos Sprague-Dawley , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/metabolismo , Testes de Toxicidade
11.
Curr Opin Drug Discov Devel ; 9(1): 84-91, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16445120

RESUMO

Predictive toxicogenomics, ie, the acquisition of advanced knowledge of the safety profile of a compound using genomic biomarkers, is a technology that provides much optimism for improving early drug discovery decisions. Toxicogenomics creates an opportunity to shift attrition to earlier stages in drug development to a point where course-corrective action can be taken with relatively lower financial costs, thus improving the efficiency of the drug development process. This review will survey the current state-of-the-art in toxicogenomics for predicting toxicity, both in vivo and in vitro, with emphasis on the use of classification algorithms and the importance of toxicogenomic databases for biomarker discovery and validation.


Assuntos
Avaliação Pré-Clínica de Medicamentos/tendências , Testes de Toxicidade/tendências , Toxicogenética/tendências , Algoritmos , Animais , Biomarcadores , Bases de Dados como Assunto , Desenho de Fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Toxicogenética/economia
12.
Am J Pharmacogenomics ; 5(3): 161-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15952870

RESUMO

The economic hurdles of drug development and the emergence of genomic technologies such as chemogenomics are combining to shift the existing paradigms in preclinical drug development. Today, the information gleaned from high content molecular data has begun to augment traditional approaches to the assessment of drug safety. The optimal approach is a hybrid strategy employing chemogenomic data and gene expression-based biomarkers of drug efficacy and toxicity to supplement low content and insensitive methods for risk assessment and mechanistic evaluation of drug candidates. Large reference databases of chemogenomic data are essential to the derivation and validation of accurate and predictive gene expression biomarkers. An example of the development of a predictive biomarker for hepatic bile duct hyperplasia is described herein. As gene expression technologies improve, biomarkers will achieve higher throughput, and become more cost effective and increasingly accurate. This will elevate the value of chemogenomics in drug development, shift attrition to earlier in the process, and reduce the overall cost of drug development. Over the past 2 to 3 years, the transition of chemogenomics from a research tool to a decision-making tool has begun and regulatory agencies are anxiously awaiting implementation of this technology to make faster and more informed evaluations of potential drugs.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Fígado/efeitos dos fármacos , Farmacogenética/métodos , Animais , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Biomarcadores , Carcinógenos/toxicidade , Bases de Dados Factuais , Perfilação da Expressão Gênica , Humanos , Hiperplasia , Técnicas In Vitro , Fígado/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oncogenes/efeitos dos fármacos , Farmacogenética/estatística & dados numéricos , Segurança
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA