Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cells ; 11(10)2022 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-35626744

RESUMO

Target modulation of the AhR for inflammatory gastrointestinal (GI) conditions holds great promise but also the potential for safety liabilities both within and beyond the GI tract. The ubiquitous expression of the AhR across mammalian tissues coupled with its role in diverse signaling pathways makes development of a "clean" AhR therapeutically challenging. Ligand promiscuity and diversity in context-specific AhR activation further complicates targeting the AhR for drug development due to limitations surrounding clinical translatability. Despite these concerns, several approaches to target the AhR have been explored such as small molecules, microbials, PROTACs, and oligonucleotide-based approaches. These various chemical modalities are not without safety liabilities and require unique de-risking strategies to parse out toxicities. Collectively, these programs can benefit from in silico and in vitro methodologies that investigate specific AhR pathway activation and have the potential to implement thresholding parameters to categorize AhR ligands as "high" or "low" risk for sustained AhR activation. Exploration into transcriptomic signatures for AhR safety assessment, incorporation of physiologically-relevant in vitro model systems, and investigation into chronic activation of the AhR by structurally diverse ligands will help address gaps in our understanding regarding AhR-dependent toxicities. Here, we review the role of the AhR within the GI tract, novel therapeutic modality approaches to target the AhR, key AhR-dependent safety liabilities, and relevant strategies that can be implemented to address drug safety concerns. Together, this review discusses the emerging therapeutic landscape of modalities targeting the AhR for inflammatory GI indications and offers a safety roadmap for AhR drug development.


Assuntos
Receptores de Hidrocarboneto Arílico , Transdução de Sinais , Animais , Trato Gastrointestinal/metabolismo , Ligantes , Mamíferos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo
2.
ALTEX ; 39(4): 560-582, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35502629

RESUMO

Drug-induced neurotoxicity is a leading cause of safety-related attrition for therapeutics in clinical trials, often driven by poor predictivity of preclinical in vitro and in vivo models of neurotoxicity. Over a dozen different iPSC-derived 3D spheroids have been described in recent years, but their ability to predict neurotoxicity in patients has not been evaluated nor compared with the predictive power of nonclinical species. To assess the predictive capabilities of human iPSC-derived neural spheroids (microBrains), we used 84 structurally diverse pharmaceuticals with robust clinical and pre-clinical datasets with varying degrees of seizurogenic and neurodegenerative liability. Drug-induced changes in neural viability and phenotypic calcium bursts were assessed using 7 endpoints based on calcium oscillation profiles and cel-lular ATP levels. These endpoints, normalized by therapeutic exposure, were used to build logistic regression models to establish endpoint cutoffs and evaluate probability for clinical neurotoxicity. The neurotoxicity score calculated from the logistic regression model could distinguish neurotoxic from non-neurotoxic clinical molecules with a specificity as high as 93.33% and a sensitivity of 53.49%, demonstrating a very low false positive rate for the prediction of seizures, convulsions, and neurodegeneration. In contrast, nonclinical species showed a higher sensitivity (75%) but much lower specificity (30.4%). The neural spheroids demonstrated higher likelihood ratio positive and inverse likelihood ratio neg-ative values compared with nonclinical safety studies. This assay has the potential to be used as a predictive assay to detect neurotoxicity in early drug discovery, aiding in the early identification of compounds that eventually may fail due to neurotoxicity.


Assuntos
Células-Tronco Pluripotentes Induzidas , Síndromes Neurotóxicas , Humanos , Síndromes Neurotóxicas/etiologia , Convulsões/induzido quimicamente , Sinalização do Cálcio , Preparações Farmacêuticas
3.
Toxicology ; 461: 152902, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34418498

RESUMO

Development of TAK-875 was discontinued when a small number of serious drug-induced liver injury (DILI) cases were observed in Phase 3 clinical trials. Subsequent studies have identified hepatocellular oxidative stress, mitochondrial dysfunction, altered bile acid homeostasis, and immune response as mechanisms of TAK-875 DILI and the contribution of genetic risk factors in oxidative response and mitochondrial pathways to the toxicity susceptibility observed in patients. We tested the hypothesis that a novel preclinical approach based on gene pathway analysis in the livers of Collaborative Cross mice could be used to identify human-relevant mechanisms of toxicity and genetic risk factors at the level of the hepatocyte as reported in a human genome-wide association study. Eight (8) male mice (4 matched pairs) from each of 45 Collaborative Cross lines were treated with a single oral (gavage) dose of either vehicle or 600 mg/kg TAK-875. As expected, liver injury was not detected histologically and few changes in plasma biomarkers of hepatotoxicity were observed. However, gene expression profiling in the liver identified hundreds of transcripts responsive to TAK-875 treatment across all strains reflecting alterations in immune response and bile acid homeostasis and the interaction of treatment and strain reflecting oxidative stress and mitochondrial dysfunction. Fold-change expression values were then used to develop pathway-based phenotypes for genetic mapping which identified candidate risk factor genes for TAK-875 toxicity susceptibility at the level of the hepatocyte. Taken together, these findings support our hypothesis that a gene pathway-based approach using Collaborative Cross mice could inform sensitive strains, human-relevant mechanisms of toxicity, and genetic risk factors for TAK-875 DILI. This novel preclinical approach may be helpful in understanding, predicting, and ultimately preventing clinical DILI for other drugs.


Assuntos
Benzofuranos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Hepatócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Sulfonas/toxicidade , Animais , Ácidos e Sais Biliares/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Camundongos de Cruzamento Colaborativo , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Hepatócitos/patologia , Humanos , Masculino , Camundongos , Fatores de Risco
4.
Nat Med ; 26(10): 1541-1548, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32895570

RESUMO

Drug-induced liver injury (DILI) is a leading cause of termination in drug development programs and removal of drugs from the market; this is partially due to the inability to identify patients who are at risk1. In this study, we developed a polygenic risk score (PRS) for DILI by aggregating effects of numerous genome-wide loci identified from previous large-scale genome-wide association studies2. The PRS predicted the susceptibility to DILI in patients treated with fasiglifam, amoxicillin-clavulanate or flucloxacillin and in primary hepatocytes and stem cell-derived organoids from multiple donors treated with over ten different drugs. Pathway analysis highlighted processes previously implicated in DILI, including unfolded protein responses and oxidative stress. In silico screening identified compounds that elicit transcriptomic signatures present in hepatocytes from individuals with elevated PRS, supporting mechanistic links and suggesting a novel screen for safety of new drug candidates. This genetic-, cellular-, organoid- and human-scale evidence underscored the polygenic architecture underlying DILI vulnerability at the level of hepatocytes, thus facilitating future mechanistic studies. Moreover, the proposed 'polygenicity-in-a-dish' strategy might potentially inform designs of safer, more efficient and robust clinical trials.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/genética , Herança Multifatorial , Polimorfismo de Nucleotídeo Único , Alelos , Benzofuranos/uso terapêutico , Estudos de Casos e Controles , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/epidemiologia , Estudos de Coortes , Conjuntos de Dados como Assunto/estatística & dados numéricos , Perfilação da Expressão Gênica , Frequência do Gene , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Análise em Microsséries , Herança Multifatorial/genética , Sulfonas/uso terapêutico
5.
Toxicol Lett ; 331: 227-234, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32522578

RESUMO

An important mechanism of chemical toxicity is the induction of oxidative stress through the production of excess reactive oxygen species (ROS). In this study, we show that the level of drug-induced ROS production between NRK52E and HepG2 cells is significantly different for several marketed drugs and a number of Takeda's internal proprietary compounds. Nifedipine, a calcium channel blocker and the initial focus of the study, was demonstrated to promote in vitro ROS production and a decrease in cell viability in NRK52E cells but not HepG2 cells. ROS production after nifedipine treatment was inhibited by a NOX inhibitor (GKT136901) but not the mitochondrial NADH dehydrogenase inhibitor, rotenone, suggesting that nifedipine decreases NRK52E cell viability primarily through a NOX-mediated pathway. To understand the breadth of NOX-mediated ROS production, 12 commercially available compounds that are structurally and/or pharmacologically related to nifedipine as well as 172 internal Takeda candidate drugs, were also evaluated against these two cell types. Over 15 % of compounds not cytotoxic to HepG2 cells (below 50 µM) were cytotoxic to NRK52E cells. Our results suggest that a combination of cell viability data from both NRK52E and HepG2 cells was superior for the prediction of in vivo toxicity findings when compared to use of only one cell line. Further, the NRK52E cell viability assay is a good predictor of NOX-mediated ROS production and can be used as a follow up assay following a negative HepG2 response to aid in the selection of suitable compounds for in vivo toxicity studies.


Assuntos
Células Epiteliais/efeitos dos fármacos , Rim/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Bioensaio , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Drogas em Investigação/toxicidade , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Hep G2 , Humanos , Concentração Inibidora 50 , Rim/metabolismo , Rim/patologia , NADPH Oxidase 4/genética , Nifedipino/toxicidade
6.
Metabolites ; 10(3)2020 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-32106514

RESUMO

Obesity is a multifactorial disease with many complications and related diseases and has become a global epidemic. To thoroughly understand the impact of obesity on whole organism homeostasis, it is helpful to utilize a systems biological approach combining gene expression and metabolomics across tissues and biofluids together with metagenomics of gut microbial diversity. Here, we present a multi-omics study on liver, muscle, adipose tissue, urine, plasma, and feces on mice fed a high-fat diet (HFD). Gene expression analyses showed alterations in genes related to lipid and energy metabolism and inflammation in liver and adipose tissue. The integration of metabolomics data across tissues and biofluids identified major differences in liver TCA cycle, where malate, succinate and oxaloacetate were found to be increased in HFD mice. This finding was supported by gene expression analysis of TCA-related enzymes in liver, where expression of malate dehydrogenase was found to be decreased. Investigations of the microbiome showed enrichment of Lachnospiraceae, Ruminococcaceae, Streptococcaceae and Lactobacillaceae in the HFD group. Our findings help elucidate how the whole organism metabolome and transcriptome are integrated and regulated during obesity.

7.
Toxicol Sci ; 167(2): 458-467, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30289550

RESUMO

TAK-875 (fasiglifam), a GPR40 agonist in development for the treatment of type 2 diabetes (T2D), was voluntarily terminated in Phase III trials due to adverse liver effects. The potential mechanisms of TAK-875 toxicity were explored by combining in vitro experiments with quantitative systems toxicology (QST) using DILIsym, a mathematical representation of drug-induced liver injury. In vitro assays revealed that bile acid transporters were inhibited by both TAK-875 and its metabolite, TAK-875-Glu. Experimental data indicated that human bile salt export pump (BSEP) inhibition by TAK-875 was mixed whereas sodium taurocholate co-transporting polypeptide (NTCP) inhibition by TAK-875 was competitive. Furthermore, experimental data demonstrated that both TAK-875 and TAK-875-Glu inhibit mitochondrial electron transport chain (ETC) enzymes. These mechanistic data were combined with a physiologically based pharmacokinetic (PBPK) model constructed within DILIsym to estimate liver exposure of TAK-875 and TAK-875-Glu. In a simulated population (SimPops) constructed to reflect T2D patients, 16/245 (6.5%) simulated individuals developed alanine aminotransferase (ALT) elevations, an incidence similar to that observed with 200 mg daily dosing in clinical trials. Determining the mode of bile acid transporter inhibition (Ki) was critical to accurate predictions. In addition, simulations conducted on a sensitive subset of individuals (SimCohorts) revealed that when either BSEP or ETC inhibition was inactive, ALT elevations were not predicted to occur, suggesting that the two mechanisms operate synergistically to produce the observed clinical response. These results demonstrate how utilizing QST methods to interpret in vitro experimental results can lead to an improved understanding of the clinically relevant mechanisms underlying drug-induced toxicity.


Assuntos
Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Benzofuranos/toxicidade , Ácidos e Sais Biliares/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Fígado/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Modelos Biológicos , Sulfonas/toxicidade , Benzofuranos/farmacocinética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Simulação por Computador , Humanos , Fígado/metabolismo , Mitocôndrias/metabolismo , Sulfonas/farmacocinética
8.
Exp Biol Med (Maywood) ; 242(16): 1579-1585, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28622731

RESUMO

Tissue chips are poised to deliver a paradigm shift in drug discovery. By emulating human physiology, these chips have the potential to increase the predictive power of preclinical modeling, which in turn will move the pharmaceutical industry closer to its aspiration of clinically relevant and ultimately animal-free drug discovery. Despite the tremendous science and innovation invested in these tissue chips, significant challenges remain to be addressed to enable their routine adoption into the industrial laboratory. This article describes the main steps that need to be taken and highlights key considerations in order to transform tissue chip technology from the hands of the innovators into those of the industrial scientists. Written by scientists from 13 pharmaceutical companies and partners at the National Institutes of Health, this article uniquely captures a consensus view on the progression strategy to facilitate and accelerate the adoption of this valuable technology. It concludes that success will be delivered by a partnership approach as well as a deep understanding of the context within which these chips will actually be used. Impact statement The rapid pace of scientific innovation in the tissue chip (TC) field requires a cohesive partnership between innovators and end users. Near term uptake of these human-relevant platforms will fill gaps in current capabilities for assessing important properties of disposition, efficacy and safety liabilities. Similarly, these platforms could support mechanistic studies which aim to resolve challenges later in development (e.g. assessing the human relevance of a liability identified in animal studies). Building confidence that novel capabilities of TCs can address real world challenges while they themselves are being developed will accelerate their application in the discovery and development of innovative medicines. This article outlines a strategic roadmap to unite innovators and end users thus making implementation smooth and rapid. With the collective contributions from multiple international pharmaceutical companies and partners at National Institutes of Health, this article should serve as an invaluable resource to the multi-disciplinary field of TC development.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Procedimentos Analíticos em Microchip/métodos , Microfluídica/métodos , Indústria Farmacêutica , Humanos , Dispositivos Lab-On-A-Chip
9.
Int J Food Sci Nutr ; 68(7): 849-864, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28276900

RESUMO

The aim was to develop novel fibres by enzymatic synthesis, to determine their total dietary fibre by AOAC method 2009.01 and to estimate their potential digestibility and assess their digestibility in vivo using glycaemic and insulinaemic responses as markers in mice and randomised clinical trial models. We found that fibre candidates to which α-(1,2) branching was added were resistant to digestion in the mouse model, depending on the amount of branching. These results show that in vivo models are needed to reliably assess the digestibility of α-glycosidic-linked oligomeric dietary fibre candidates, possibly due to absence of brush border α-glucosidase activity in the current in vitro assessment. α-(1,3)-linked and α-(1,6)-linked glucose oligomers were completely digested in humans and mice. In conclusion, it is possible to develop dietary soluble fibres by enzymatic synthesis. Adding α-(1,2) branching increases their resistance to digestion in vivo and can thus improve their suitability as potential fibre candidates. Clinical Trial Registry: ClinicalTrials.gov, NCT02701270.


Assuntos
Fibras na Dieta/análise , Fibras na Dieta/metabolismo , Digestão/fisiologia , Adulto , Animais , Área Sob a Curva , Bactérias/metabolismo , Glicemia/efeitos dos fármacos , Glicemia/fisiologia , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade
10.
Toxicol Sci ; 157(1): 50-61, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28108665

RESUMO

Fasiglifam (TAK-875), a Free Fatty Acid Receptor 1 (FFAR1) agonist in development for the treatment of type 2 diabetes, was voluntarily terminated in phase 3 due to adverse liver effects. A mechanistic investigation described in this manuscript focused on the inhibition of bile acid (BA) transporters as a driver of the liver findings. TAK-875 was an in vitro inhibitor of multiple influx (NTCP and OATPs) and efflux (BSEP and MRPs) hepatobiliary BA transporters at micromolar concentrations. Repeat dose studies determined that TAK-875 caused a dose-dependent increase in serum total BA in rats and dogs. Additionally, there were dose-dependent increases in both unconjugated and conjugated individual BAs in both species. Rats had an increase in serum markers of liver injury without correlative microscopic signs of tissue damage. Two of 6 dogs that received the highest dose of TAK-875 developed liver injury with clinical pathology changes, and by microscopic analysis had portal granulomatous inflammation with neutrophils around a crystalline deposition. The BA composition of dog bile also significantly changed in a dose-dependent manner following TAK-875 administration. At the highest dose, levels of taurocholic acid were 50% greater than in controls with a corresponding 50% decrease in taurochenodeoxycholic acid. Transporter inhibition by TAK-875 may cause liver injury in dogs through altered bile BA composition characteristics, as evidenced by crystalline deposition, likely composed of test article, in the bile duct. In conclusion, a combination of in vitro and in vivo evidence suggests that BA transporter inhibition could contribute to TAK-875-mediated liver injury in dogs.


Assuntos
Benzofuranos/toxicidade , Ácidos e Sais Biliares/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Homeostase/efeitos dos fármacos , Sulfonas/toxicidade , Administração Oral , Animais , Benzofuranos/administração & dosagem , Benzofuranos/farmacocinética , Células Cultivadas , Cães , Relação Dose-Resposta a Droga , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Sulfonas/administração & dosagem , Sulfonas/farmacocinética
11.
Toxicol Appl Pharmacol ; 289(3): 560-72, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26450648

RESUMO

Doxorubicin (DOX) is a potent and effective broad-spectrum anthracycline antitumor agent, but its clinical usefulness is restricted by cardiotoxicity. This study compared pharmacokinetic, functional, structural and biochemical effects of single dose DOX bolus or 3-h continuous iv infusion (3-h iv) in the Han­Wistar rat to characterize possible treatment-related differences in drug safety over a 72 h observation period. Both DOX dosing paradigms significantly altered blood pressure, core body temperature and QA interval (indirect measure of cardiac contractility); however, there was no recovery observed in the bolus iv treatment group. Following the 3-h iv treatment, blood pressures and QA interval normalized by 36 h then rose above baseline levels over 72 h. Both treatments induced biphasic changes in heart rate with initial increases followed by sustained decreases. Cardiac injury biomarkers in plasma were elevated only in the bolus iv treatment group. Tissue cardiac injury biomarkers, cardiac mitochondrial complexes I, III and V and cardiac mitochondrial sphingolipids were decreased only in the bolus iv treatment group. Results indicate that each DOX dosing paradigm deregulates sinus rhythm.However, slowing the rate of infusion allows for functional compensation of blood pressure and may decrease the likelihood of cardiac myocyte necrosis via a mechanism associated with reduced mitochondrial damage.


Assuntos
Doxorrubicina/administração & dosagem , Coração/efeitos dos fármacos , Rim/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Esfingolipídeos/metabolismo , Administração Intravenosa/métodos , Animais , Biomarcadores/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Esquema de Medicação , Coração/fisiopatologia , Rim/metabolismo , Rim/patologia , Ratos , Ratos Wistar
13.
Toxicol Sci ; 137(2): 278-91, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24189134

RESUMO

Despite six decades of clinical experience with the polymyxin class of antibiotics, their dose-limiting nephrotoxicity remains difficult to predict due to a paucity of sensitive biomarkers. Here, we evaluate the performance of standard of care and next-generation biomarkers of renal injury in the detection and monitoring of polymyxin-induced acute kidney injury in male Han Wistar rats using colistin (polymyxin E) and a polymyxin B (PMB) derivative with reduced nephrotoxicity, PMB nonapeptide (PMBN). This study provides the first histopathological and biomarker analysis of PMBN, an important test of the hypothesis that fatty acid modifications and charge reductions in polymyxins can reduce their nephrotoxicity. The results indicate that alterations in a panel of urinary kidney injury biomarkers can be used to monitor histopathological injury, with Kim-1 and α-GST emerging as the most sensitive biomarkers outperforming clinical standards of care, serum or plasma creatinine and blood urea nitrogen. To enable the prediction of polymyxin-induced nephrotoxicity, an in vitro cytotoxicity assay was employed using human proximal tubule epithelial cells (HK-2). Cytotoxicity data in these HK-2 cells correlated with the renal toxicity detected via safety biomarker data and histopathological evaluation, suggesting that in vitro and in vivo methods can be incorporated within a screening cascade to prioritize polymyxin class analogs with more favorable renal toxicity profiles.


Assuntos
Antibacterianos/toxicidade , Colistina/toxicidade , Nefropatias/urina , Polimixina B/análogos & derivados , Animais , Antibacterianos/administração & dosagem , Antibacterianos/farmacocinética , Biomarcadores/urina , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colistina/administração & dosagem , Colistina/farmacocinética , Interpretação Estatística de Dados , Relação Dose-Resposta a Droga , Diagnóstico Precoce , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Polimixina B/administração & dosagem , Polimixina B/farmacocinética , Polimixina B/toxicidade , Prognóstico , Ratos , Ratos Wistar
14.
Toxicol Sci ; 135(2): 402-13, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23897988

RESUMO

Cardiovascular (CV) toxicity is a leading contributor to drug attrition. Implementing earlier testing has successfully reduced human Ether-à-go-go-Related Gene-related arrhythmias. How- ever, analogous assays targeting functional CV effects remain elusive. Demand to address this gap is particularly acute for kinase inhibitors (KIs) that suffer frequent CV toxicity. The drug class also presents some particularly challenging requirements for assessing functional CV toxicity. Specifically, an assay must sense a downstream response that integrates diverse kinase signaling pathways. In addition, sufficient throughput is essential for handling inherent KI nonselectivity. A new opportunity has emerged with cellular impedance technology, which detects spontaneous beating cardiomyocytes. Impedance assays sense morphology changes downstream of cardiomyocyte contraction. To evaluate cardiomyocyte impedance assays for KI screening, we investigated two distinct KI classes where CV toxicity was discovered late and target risks remain unresolved. Microtubule-associated protein/microtubule affinity regulating kinase (MARK) inhibitors decrease blood pressure in dogs, whereas checkpoint kinase (Chk) inhibitors (AZD7762, SCH900776) exhibit dose-limiting CV toxicities in clinical trials. These in vivo effects manifested in vitro as cardiomyocyte beat cessation. MARK effects were deemed mechanism associated because beat inhibition potencies correlated with kinase inhibition, and gene knockdown and microtubule-targeting agents suppressed beating. MARK inhibitor impedance and kinase potencies aligned with rat blood pressure effects. Chk inhibitor effects were judged off-target because Chk and beat inhibition potencies did not correlate and knockdowns did not alter beating. Taken together, the data demonstrate that cardiomyocyte impedance assays can address three unmet needs-detecting KI functional cardiotoxicity in vitro, determining mechanism of action, and supporting safety structure-activity relationships.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Inibidores de Proteínas Quinases/farmacologia , Animais , Cães , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ratos , Ratos Wistar
15.
Toxicol Lett ; 219(1): 49-58, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23470867

RESUMO

Predicting human safety risks of novel xenobiotics remains a major challenge, partly due to the limited availability of human cells to evaluate tissue-specific toxicity. Recent progress in the production of human induced pluripotent stem cells (hiPSCs) may fill this gap. hiPSCs can be continuously expanded in culture in an undifferentiated state and then differentiated to form most cell types. Thus, it is becoming technically feasible to generate large quantities of human cell types and, in combination with relatively new detection methods, to develop higher-throughput in vitro assays that quantify tissue-specific biological properties. Indeed, the first wave of large scale hiSC-differentiated cell types including patient-derived hiPSCS are now commercially available. However, significant improvements in hiPSC production and differentiation processes are required before cell-based toxicity assays that accurately reflect mature tissue phenotypes can be delivered and implemented in a cost-effective manner. In this review, we discuss the promising alignment of hiPSCs and recently emerging technologies to quantify tissue-specific functions. We emphasize liver, cardiovascular, and CNS safety risks and highlight limitations that must be overcome before routine screening for toxicity pathways in hiSC-derived cells can be established.


Assuntos
Descoberta de Drogas/métodos , Células-Tronco Pluripotentes Induzidas , Testes de Toxicidade/métodos , Técnicas de Cultura de Células , Sistema Nervoso Central/citologia , Sistema Nervoso Central/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Fígado/citologia , Fígado/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Medicina Regenerativa/tendências
16.
Assay Drug Dev Technol ; 10(6): 525-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22574652

RESUMO

Cardiovascular toxicity is a leading contributor to drug withdrawal and late-stage attrition. Earlier and broader screening is a validated approach to build-in cardiovascular safety as demonstrated with human Ether-à-go-go-related gene (hERG) screening to reduce drug-induced arrhythmia. There is an urgent need for novel in vitro assays to address other mechanistic aspects of cardiovascular function, including contractility, heart rate, toxicity, hypertrophy, and non-hERG arrhythmia. Recent advances in label-free cellular impedance technology now enable tracking of spontaneous, synchronized beating of cultured cardiomyocytes. Analysis of beating allows integrated detection that is downstream of electrical and mechanical aspects of contraction. Here, we evaluate impedance-based cardiomyocyte responses against criteria required for drug screening. The throughput and sensitivity allowed for rapid assay development. Critical variables for rat neonatal cardiomyocyte assays included cell density and serum levels. Once optimized, consistent, stable beating for at least 3 days was straight-forward to achieve. In tests of compounds spanning a breadth of target classes, the potency values showed excellent precision, wide dynamic range, and consistency across multiple experiments. Cardiomyocyte impedance assays can extract multiple beat-related parameters. In these experiments, rate, amplitude, and rise slope were examined and each yielded acceptable precision. Potency values calculated by beat rate and amplitude were highly correlated for most compounds although selected compounds displayed unique profiles indicative of different mechanisms. Tests with known cardiovascular active drugs revealed concordance with clinical findings. Thus, impedance assays combine novel features including sensitivity to contractile activity, versatile data analysis, and robust/translatable data in a format with sufficient throughput to become a valuable addition to the cardiovascular in vitro screening arsenal.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Animais , Animais Recém-Nascidos , Bioensaio/métodos , Fármacos Cardiovasculares/farmacologia , Células Cultivadas , Meios de Cultura , Interpretação Estatística de Dados , Dimetil Sulfóxido/farmacologia , Impedância Elétrica , Canais de Potássio Éter-A-Go-Go/genética , Frequência Cardíaca/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Relação Estrutura-Atividade
17.
Toxicol Sci ; 116(2): 375-81, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20430866

RESUMO

The International Life Sciences Institute, Health and Environmental Sciences Institute sponsored a workshop entitled "State of the Science: Evaluating Epigenetic Changes," hosted by the National Institute of Environmental Health Sciences, Research Triangle Park, NC, 28-30 October 2009. The goal was to evaluate and enhance the scientific knowledge base regarding epigenetics and its role in disease, including potential relationships between epigenetic changes and transgenerational effects. A distinguishing aspect of the workshop was the highly interactive discussion session on the final morning. Meeting participants formed breakout groups (with representation from academia, industry, and government in each group) and were tasked with integrating their previous knowledge of epigenetics with what was learned during the workshop. The participants addressed the issue of what needs to be known prior to thinking about incorporating an epigenetic evaluation into safety assessment. To this end, the breakout groups were asked to address the following questions: (1) What model systems might be employed to evaluate the ability of a chemical to produce an epigenetic change (affecting the F1 and/or F3 generation); (2) What end points/targets might be evaluated; (3) What techniques might be employed; and (4) Regulatory Perspective: When is it appropriate to incorporate "new" science, in this case epigenetics, into the regulatory process? What does one need to know, what are the pitfalls and how might these be overcome/avoided? The basis of this paper is a synopsis of these discussions. The workshop highlighted the fact that the field of epigenetics is evolving at a very rapid pace and indicated that a great deal needs to be learned prior to being able to rationally incorporate an epigenetic evaluation into safety assessment. The value of the workshop is that it called attention to key data/knowledge gaps that should serve to focus attention on the areas where research and new thinking are needed to better understand epigenetics and its relationship to safety assessment.


Assuntos
Epigênese Genética/efeitos dos fármacos , Medição de Risco , Animais , Metilação de DNA , Determinação de Ponto Final , Humanos , Saúde Pública
18.
J Comput Biol ; 16(12): 1671-88, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20047490

RESUMO

The rapid advances in proteomic analyses coupled with the completion of multiple genomes have led to an increased demand for determining protein functions. The first step is classification or prediction into families. A method was developed for the prediction of protein family based only on protein sequence using support vector machine (SVM) models. In these models, the amino acids were classified into three categories (apolar, polar, and charged). Consecutive fragments ranging from one to five were annotated by amino acid type to define the protein features of each protein. SVM models were constructed based on the protein features of a training set of proteins and then examined with an independent set of proteins. The approach was tested for 20 protein families from the iProClass database of Protein Information Resources (PIR). For two-class SVM models, an average prediction accuracy of 0.9985 was achieved, while for multi-class SVM models an accuracy of 0.9941 was achieved. This study demonstrates that SVM based methods can accurately recognize and predict the protein family to which a sequence belongs based solely on its primary amino acid sequence.


Assuntos
Família Multigênica , Proteínas/química , Proteínas/classificação , Análise de Sequência de Proteína/métodos , Algoritmos , Sequência de Aminoácidos , Bases de Dados de Proteínas , Modelos Teóricos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
19.
Toxicol Appl Pharmacol ; 235(1): 25-32, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19103213

RESUMO

Drug-induced liver injury has been associated with the generation of reactive metabolites, which are primarily detoxified via glutathione conjugation. In this study, it was hypothesized that molecules involved in the synthesis of glutathione would be diminished to replenish the glutathione depleted through conjugation reactions. Since S-adenosylmethionine (SAMe) is the primary source of the sulfur atom in glutathione, UPLC/MS and NMR were used to evaluate metabolites involved with the transulfuration pathway in urine samples collected during studies of eight liver toxic compounds in Sprague-Dawley rats. Urinary levels of creatine were increased on day 1 or day 2 in 8 high dose liver toxicity studies. Taurine concentration in urine was increased in only 3 of 8 liver toxicity studies while SAMe was found to be reduced in 4 of 5 liver toxicity studies. To further validate the results from the metabonomic studies, microarray data from rat liver samples following treatment with acetaminophen was obtained from the Gene Expression Omnibus (GEO) database. Some genes involved in the trans-sulfuration pathway, including guanidinoacetate N-methyltransferase, glycine N-methyltransferase, betaine-homocysteine methyltransferase and cysteine dioxygenase were found to be significantly decreased while methionine adenosyl transferase II, alpha increased at 24 h post-dosing, which is consistent with the SAMe and creatine findings. The metabolic and transcriptomic results show that the trans-sulfuration pathway from SAMe to glutathione was disturbed due to the administration of heptatotoxicants.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Fígado/efeitos dos fármacos , Animais , Ensaio Cometa , Creatina/metabolismo , Creatina/urina , Perfilação da Expressão Gênica , Glutationa/metabolismo , Fígado/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley , S-Adenosilmetionina/análogos & derivados , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/urina , Taurina/metabolismo , Taurina/urina
20.
Artigo em Inglês | MEDLINE | ID: mdl-18472313

RESUMO

Urinary metabolic perturbations associated with acute and chronic acetaminophen-induced hepatotoxicity were investigated using nuclear magnetic resonance (NMR) spectroscopy and ultra performance liquid chromatography/mass spectrometry (UPLC/MS) metabonomics approaches to determine biomarkers of hepatotoxicity. Acute and chronic doses of acetaminophen (APAP) were administered to male Sprague-Dawley rats. NMR and UPLC/MS were able to detect both drug metabolites and endogenous metabolites simultaneously. The principal component analysis (PCA) of NMR or UPLC/MS spectra showed that metabolic changes observed in both acute and chronic dosing of acetaminophen were similar. Histopathology and clinical chemistry studies were performed and correlated well with the PCA analysis and magnitude of metabolite changes. Depletion of antioxidants (e.g. ferulic acid), trigonelline, S-adenosyl-L-methionine, and energy-related metabolites indicated that oxidative stress was caused by acute and chronic acetaminophen administration. Similar patterns of metabolic changes in response to acute or chronic dosing suggest similar detoxification and recovery mechanisms following APAP administration.


Assuntos
Acetaminofen/intoxicação , Doença Hepática Induzida por Substâncias e Drogas , Cromatografia Líquida de Alta Pressão/métodos , Biologia Computacional/métodos , Hepatopatias/urina , Espectrometria de Massas/métodos , Metabolismo , Ressonância Magnética Nuclear Biomolecular/métodos , Ratos Sprague-Dawley/urina , Animais , Fígado/patologia , Hepatopatias/patologia , Masculino , Necrose/patologia , Necrose/urina , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA