Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharmacol Toxicol Methods ; 105: 106897, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32663523

RESUMEN

INTRODUCTION: Alterations in cardiac contractility can have significant clinical implications, highlighting the need for early detection of potential liabilities. Pre-clinical methods to assess contractility are typically invasive and their translation to human measures of cardiac function are not well defined. Clinically, cardiac function is most often measured non-invasively using echocardiography. The objective of these studies was to introduce echocardiography into standard large animal cardiovascular safety pharmacology studies and determine the feasibility of this combination. METHODS: A consortia of laboratories combined their data sets for evaluation. At each site, telemetered beagle dogs, in a 4 × 4 Latin square crossover study design (n = 4), were administered either pimobendan (positive inotrope) or atenolol (negative inotrope) orally at clinically relevant dose levels. Standard telemetry parameters were collected (heart rate, mean arterial blood pressure, etc.) continuously over 24 h, as well as derived contractility endpoints: QA interval and LV +dP/dtmax. At Tmax, echocardiography was performed in conscious dogs with minimal restraint to collect contractility parameters: ejection fraction (EF) and fractional shortening (FS). RESULTS: Correlations between telemetry and echo contractility endpoints showed that, in general, a change in LV +dP/dtmax of 1000 mmHg/s translates to a 5.2% change in EF and a 4.2% change in FS. Poor correlations were shown between QA interval derived simultaneously, to both EF and FS. DISCUSSION: Comparing data from telemetry-only groups to those that included echocardiography collections showed no effect in the ability to interpret test article-related effects, providing the foundation for the inclusion of echocardiography without compromising standard telemetry data quality.


Asunto(s)
Cardiotónicos/efectos adversos , Hemodinámica/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Animales , Atenolol/efectos adversos , Presión Sanguínea/efectos de los fármacos , Estudios Cruzados , Perros , Ecocardiografía/métodos , Electrocardiografía/métodos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Piridazinas/efectos adversos , Telemetría/métodos , Función Ventricular Izquierda/efectos de los fármacos
2.
Artículo en Inglés | MEDLINE | ID: mdl-29042254

RESUMEN

INTRODUCTION: Cardiac sodium channel antagonists have historically been used to treat cardiac arrhythmias by preventing the reentry of the electrical impulse that could occur following myocardial damage. However, clinical studies have highlighted a significant increase in mortality associated with such treatment. Cardiac sodium channel antagonist activity is now seen as an off-target pharmacology that should be mitigated during the drug development process. The aim of this study was to examine the correlation between in vitro/ex vivo assays that are routinely used to measure Nav1.5 activity and determine the translatability of the individual assays to QRS prolongation in the clinic. METHODS: A set of clinical compounds with known Nav1.5 activity was profiled in several in vitro/ex vivo assays (binding, membrane potential, patch clamp and the Langendorff isolated heart). Clinical data comprising compound exposure levels and changes in QRS interval were obtained from the literature. Sensitivity/specificity analysis was performed with respect to the clinical outcome. RESULTS: The in vitro assays showed utility in predicting QRS prolongation in the clinic. Optimal thresholds were defined for each assay (binding: IC20; membrane potential: IC10; patch clamp: IC20) and sensitivity (69-88%) and specificity (53-84%) values were shown to be similar between assay formats. DISCUSSION: The data provide clear statistical insight into the translatability of Nav1.5 antagonism data generated in vitro to potential clinical outcomes. These results improve our ability to understand the liability posed by such activity in novel development compounds at an early stage.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Contracción Miocárdica/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Arritmias Cardíacas/diagnóstico , Células CHO , Cricetinae , Cricetulus , Perros , Evaluación Preclínica de Medicamentos/métodos , Electrocardiografía , Cobayas , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Masculino , Sensibilidad y Especificidad , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
3.
Cell Metab ; 25(5): 1147-1159.e10, 2017 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467931

RESUMEN

The AMP-activated protein kinase (AMPK) is a potential therapeutic target for metabolic diseases based on its reported actions in the liver and skeletal muscle. We evaluated two distinct direct activators of AMPK: a non-selective activator of all AMPK complexes, PF-739, and an activator selective for AMPK ß1-containing complexes, PF-249. In cells and animals, both compounds were effective at activating AMPK in hepatocytes, but only PF-739 was capable of activating AMPK in skeletal muscle. In diabetic mice, PF-739, but not PF-249, caused a rapid lowering of plasma glucose levels that was diminished in the absence of skeletal muscle, but not liver, AMPK heterotrimers and was the result of an increase in systemic glucose disposal with no impact on hepatic glucose production. Studies of PF-739 in cynomolgus monkeys confirmed translation of the glucose lowering and established activation of AMPK in skeletal muscle as a potential therapeutic approach to treat diabetic patients.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminopiridinas/farmacología , Activadores de Enzimas/farmacología , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Indoles/farmacología , Aminopiridinas/uso terapéutico , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/uso terapéutico , Femenino , Hipoglucemiantes/uso terapéutico , Indoles/uso terapéutico , Hígado/efectos de los fármacos , Hígado/metabolismo , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo
4.
J Med Chem ; 59(13): 6313-28, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27275946

RESUMEN

It is hypothesized that selective muscarinic M1 subtype activation could be a strategy to provide cognitive benefits to schizophrenia and Alzheimer's disease patients while minimizing the cholinergic side effects observed with nonselective muscarinic orthosteric agonists. Selective activation of M1 with a positive allosteric modulator (PAM) has emerged as a new approach to achieve selective M1 activation. This manuscript describes the development of a series of M1-selective pyridone and pyridine amides and their key pharmacophores. Compound 38 (PF-06767832) is a high quality M1 selective PAM that has well-aligned physicochemical properties, good brain penetration and pharmacokinetic properties. Extensive safety profiling suggested that despite being devoid of mAChR M2/M3 subtype activity, compound 38 still carries gastrointestinal and cardiovascular side effects. These data provide strong evidence that M1 activation contributes to the cholinergic liabilities that were previously attributed to activation of the M2 and M3 receptors.


Asunto(s)
Descubrimiento de Drogas , Ácidos Picolínicos/farmacología , Receptor Muscarínico M1/agonistas , Tiazoles/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Modelos Moleculares , Estructura Molecular , Ácidos Picolínicos/síntesis química , Ácidos Picolínicos/química , Ratas , Receptor Muscarínico M1/metabolismo , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/química
5.
Artículo en Inglés | MEDLINE | ID: mdl-27039257

RESUMEN

INTRODUCTION: The Safety Pharmacology Society (SPS) and National Centre for the Replacement, Refinement & Reduction of Animals in Research (NC3Rs) conducted a survey and workshop in 2015 to define current industry practices relating to housing of non-rodents during telemetry recordings in safety pharmacology and toxicology studies. The aim was to share experiences, canvas opinion on the study procedures/designs that could be used and explore the barriers to social housing. METHODS: Thirty-nine sites, either running studies (Sponsors or Contract Research Organisations, CROs) and/or outsourcing work responded to the survey (51% from Europe; 41% from USA). RESULTS: During safety pharmacology studies, 84, 67 and 100% of respondents socially house dogs, minipigs and non-human primates (NHPs) respectively on non-recording days. However, on recording days 20, 20 and 33% of respondents socially house the animals, respectively. The main barriers for social housing were limitations in the recording equipment used, study design and animal temperament/activity. During toxicology studies, 94, 100 and 100% of respondents socially house dogs, minipigs and NHPs respectively on non-recording days. However, on recording days 31, 25 and 50% of respondents socially house the animals, respectively. The main barriers for social housing were risk of damage to and limitations in the recording equipment used, food consumption recording and temperament/activity of the animals. CONCLUSIONS: Although the majority of the industry does not yet socially house animals during telemetry recordings in safety pharmacology and toxicology studies, there is support to implement this refinement. Continued discussions, sharing of best practice and data from companies already socially housing, combined with technology improvements and investments in infrastructure are required to maintain the forward momentum of this refinement across the industry.


Asunto(s)
Hemodinámica/efectos de los fármacos , Vivienda para Animales , Medio Social , Animales , Perros , Evaluación Preclínica de Medicamentos , Farmacología/métodos , Primates , Seguridad , Encuestas y Cuestionarios , Porcinos , Porcinos Enanos , Telemetría , Temperamento , Toxicología/métodos
6.
Artículo en Inglés | MEDLINE | ID: mdl-26844408

RESUMEN

INTRODUCTION: The cardiovascular liability of candidate compounds can be evaluated by a number of methods including implanted telemetry, jacketed telemetry and surface lead electrocardiogram (ECG). The utility of the new PhysioTel™ Digital M11 cardiovascular telemetry implant was evaluated in monkeys and dogs. METHODS: Eight monkeys and dogs (4 males and 4 females per species) were implanted with the M11 device utilizing a femoral blood pressure catheter and periosteal ECG leads. The signal quality of the ECGs was determined as a percentage of software-matched waveforms and as a percentage of signal loss during the recording periods. To investigate sensitivity for detecting changes in QT/QTc and HR/BP, moxifloxacin and doxazosin were administered to monkeys and dogs implanted with the M11 device. Additionally, histopathological evaluation of the implant site was completed. RESULTS: For both monkey and dog, the percentage of recognizable waveforms was high (65% and 85%, respectively), while the average amount of signal loss was low (1% and 3%, respectively), indicating that the M11 implants delivered data of sufficient quality. In monkeys, moxifloxacin (90mg/kg) induced QT and QTc prolongation up to 22 and 12ms, respectively, while at 30mg/kg in dogs, the maximal increases in QT and QTc were 13 and 16ms, respectively. Doxazosin (1.5 and 1.0mg/kg) produced HR increases up to 35 and 29bpm with decreases in blood pressure up to -14 and -26mmHg in monkeys and dogs, respectively. The histopathological impact of the implant, catheter and biopotential leads was limited to expected minor local inflammatory changes as assessed at necropsy and with microscopic examination. DISCUSSION: Based upon the results of this study, the PhysioTel™ Digital M11 is a suitable technology for assessing cardiovascular parameters in monkeys and dogs, and because of the size and limited invasiveness of the implant, is well positioned for use on toxicology studies.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Síndrome de QT Prolongado/fisiopatología , Telemetría/métodos , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Sistema Cardiovascular/efectos de los fármacos , Perros , Doxazosina/farmacología , Electrocardiografía/métodos , Femenino , Fluoroquinolonas/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Síndrome de QT Prolongado/tratamiento farmacológico , Macaca fascicularis , Masculino , Moxifloxacino
7.
J Pharmacol Toxicol Methods ; 54(3): 261-72, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16564186

RESUMEN

INTRODUCTION: A growing number of drugs have reportedly been associated with delayed ventricular repolarization and a potentially fatal but rare arrhythmia, torsade de pointes (TdP). There is obviously a call for a validated proarrhythmia model that distinguishes proarrhythmic drugs from nonarrhythmogenic drugs. METHODS: In this article, we validated the arterially perfused rabbit left ventricular wedge preparation model and examined its use in predicting proarrhythmic potentials of drugs. A fairly detailed methodological description about this technically challenging model was given, aiming to help others establish the assay successfully. Parameters commonly used in the action potential studies were verified and critical experimental conditions (e.g. stability and reproducibility of recordings) were examined. Six commercially available compounds with various proarrhythmic potentials were administered in the model to evaluate their correlations with individual clinical outcomes. RESULTS: Our study indicated that, in a successful experiment, the action potential duration (APD) can be stably maintained for several hours without intervention. Dofetilide, DL-sotalol, cisapride, risperidone and moxifloxacin increased endo- and epicardial APD(90), QT interval and T(P-E) (peak-to-end time of the T wave) in a reverse use-dependent manner within clinically relevant concentration ranges. Phase 2 early afterdepolarizations (EADs) were observed at 1.6, 2.3, 16.7, 37.5 and 7.9 fold, respectively, their corresponding unbound therapeutic concentrations. In contrast, fluoxetine at up to 3 microM (approximately 35 fold unbound therapeutic mean plasma concentration after 60 mg/day, p.o. for 5 weeks) had only a mild prolonging effect on APD(90) and QT with essentially no effect on T(P-E). DISCUSSION: Our results strongly support the usefulness of this model in predicting a compound's arrhythmogenic potential in humans within clinically relevant concentration ranges, and the experimental results with this model need to be interpreted in light of each drug's pharmacokinetic and pharmacodynamic behavior in clinic.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Conejos , Taquicardia Ventricular/inducido químicamente , Potenciales de Acción/efectos de los fármacos , Animales , Vasos Coronarios , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Electrocardiografía , Femenino , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Técnicas In Vitro , Perfusión , Taquicardia Ventricular/fisiopatología
8.
Mol Pharmacol ; 68(3): 876-84, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15976038

RESUMEN

A variety of drugs has been reported to cause acquired long QT syndrome through inhibition of the IKr channel. Screening compounds in early discovery and development stages against their ability to inhibit IKr or the hERG channel has therefore become an indispensable procedure in the pharmaceutical industry. In contrast to numerous hERG channel blockers discovered during screening, only (3R,4R)-4-[3-(6-methoxyquinolin-4-yl)-3-oxo-propyl]-1-[3-(2,3,5-trifluoro-phenyl)-prop-2-ynyl]-piperidine-3-carboxylic acid (RPR260243) has been reported so far to enhance the hERG current. In this article, we describe several potent mechanistically distinct hERG channel enhancers. One example is PD-118057 (2-{4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid) which produced average increases of 5.5 +/- 1.1, 44.8 +/- 3.1, and 111.1 +/- 21.7% in the peak tail hERG current at 1, 3, and 10 muM, respectively, in human embryonic kidney 293 cells. PD-118057 did not affect the voltage dependence and kinetics of gating parameters, nor did it require open conformation of the channel. In isolated guinea pig cardiomyocytes, PD-118057 showed no major effect on I(Na), I(Ca,L), I(K1), and I(Ks). PD-118057 shortened the action potential duration and QT interval in arterially perfused rabbit ventricular wedge preparation in a concentration-dependent manner. The presence of 3 muM PD-118057 prevented action potential duration and QT prolongation caused by dofetilide. "Early after-depolarizations" induced by dofetilide were also completely eliminated by 3 microM PD-118057. Although further investigation is warranted to evaluate the therapeutic value and safety profile of these compounds, our data support the notion that hERG activation by pharmaceuticals may offer a new approach in the treatment of delayed repolarization conditions, which may occur in patients with inherited or acquired long QT syndrome, congestive heart failure, and diabetes.


Asunto(s)
Antiarrítmicos/farmacología , Piperidinas/farmacología , Canales de Potasio con Entrada de Voltaje/agonistas , Quinolinas/farmacología , ortoaminobenzoatos/farmacología , Animales , Células Cultivadas , Clorobencenos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Cobayas , Corazón/efectos de los fármacos , Humanos , Masculino , Fenetilaminas/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/fisiología , Sulfonamidas/farmacología
9.
Br J Pharmacol ; 145(1): 15-23, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15711592

RESUMEN

Pentamidine, an antiprotozoal agent, has been traditionally known to cause QT prolongation and arrhythmias; however, its ionic mechanism has not been illustrated. In a stable HEK-293 cell line, we observed a concentration-dependent inhibition of the hERG current with an IC50 of 252 microM. In freshly isolated guinea-pig ventricular myocytes, pentamidine showed no effect on the L-type calcium current at concentrations up to 300 microM, with a slight prolongation of the action potential duration at this concentration. Since the effective concentrations of pentamidine on the hERG channel and APD were much higher than clinically relevant exposures (approximately 1 microM free or lower), we speculated that this drug might not prolong the QT interval through direct inhibition of I(Kr) channel. We therefore incubated hERG-HEK cells in 1 and 10 microM pentamidine-containing media (supplemented with 10% serum) for 48 h, and examined the hERG current densities in the vehicle control and pentamidine-treated cells. In all, 36 and 85% reductions of the current densities were caused by 1- and 10-microM pentamidine treatment (P<0.001 vs control), respectively. A similar level of reduction of the hERG polypeptides and a reduced intensity of the hERG protein on the surface membrane in treated cells were observed by Western blot analysis and laser-scanning confocal microscopy, respectively. Taken together, our data imply that chronic administration of pentamidine at clinically relevant exposure reduces the membrane expression of the hERG channel, which may most likely be the major mechanism of QT prolongation and torsade de pointes reported in man.


Asunto(s)
Antiprotozoarios/farmacología , Expresión Génica/efectos de los fármacos , Pentamidina/farmacología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Animales , Línea Celular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Cobayas , Humanos , Síndrome de QT Prolongado/inducido químicamente , Masculino , Miocardio/metabolismo , Canales de Potasio con Entrada de Voltaje/biosíntesis
10.
J Mol Cell Cardiol ; 37(5): 1031-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15522280

RESUMEN

Various drugs are reported to prolong the QT-interval on the surface ECG, thereby increasing the risk of developing a potentially fatal arrhythmia known as Torsades de Pointes (TdP). TdP case reports for these drugs have often been associated with risk factors such as overdosing, concomitant drugs and/or existing pathophysiological conditions. A few cases appear to be devoid of these factors. To determine what role genetic variation in the hERG gene plays in drug-induced arrhythmias, we screened DNA samples collected from 105 atrial-fibrillation patients treated with dofetilide for polymorphisms, seven of whom developed TdP. An uncommon missense change, R1047L, was identified in two of seven patients who experienced TdP as compared with five of 98 individuals who were free of TdP. Included in the affected individuals was the only subject homozygous for this SNP. Cellular electrophysiological studies revealed a 10-mV positive shift in the steady-state activation curve of the 1047L hERG channel stably expressed in HEK-293 cells as compared with the wild-type (WT) channel. The activation and inactivation kinetics of the 1047L current were significantly slower than the WT (P < 0.05) at given membrane potentials. A computer simulation using a rabbit ventricular myocyte model indicated that same extent of changes in the I(Kr) channel may result in an approximately 15% prolongation in the action potential duration. Our study suggests that 1047L leads to a functional impairment of the hERG channel, which may contribute to the higher incidence of TdP in 1047L carriers when challenged with a channel blocker.


Asunto(s)
Antiarrítmicos/efectos adversos , Fenetilaminas/efectos adversos , Polimorfismo de Nucleótido Simple/genética , Canales de Potasio con Entrada de Voltaje/genética , Sulfonamidas/efectos adversos , Torsades de Pointes/inducido químicamente , Torsades de Pointes/genética , Animales , Arginina/genética , Línea Celular , Simulación por Computador , Canal de Potasio ERG1 , Electrofisiología/métodos , Canales de Potasio Éter-A-Go-Go , Homocigoto , Humanos , Leucina/genética , Mutación Missense/genética , Técnicas de Placa-Clamp , Conejos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...