Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Assay Drug Dev Technol ; 11(9-10): 544-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24147984

RESUMEN

The human Ether-à-go-go related gene (hERG) potassium channel has been widely used to counter screen potential pharmaceuticals as a biomarker to predict clinical QT prolongation. Thus, higher throughput assays of hERG are valuable for early in vitro screening of drug candidates to minimize failure in later-stage drug development due to this potentially adverse cardiac risk. We have developed a novel method utilizing potassium fluoride to improve throughput of hERG counter screening with an automated patch clamp system, PatchXpress 7000A. In that method, ∼50% substitution of internal Cl(-) with F(-) greatly increases success rate without substantially altering the biophysical properties of the hERG channel or compromising data quality. However, effect of F(-) or other halide ions on hERG channel properties has not been studied in detail. In this study, we examined effects of complete replacement of Cl(-) in internal solution with halide ions, F(-), or Br(-). We found that (1) F(-) slightly shifts the voltage dependence of hERG channel activation to more positive voltages, while Br(-) shifts it to more negative voltages; (2) Br(-) shifts to more positive voltages both the inactivation-voltage relationship and the peak position of channel full activation of hERG; (3) F(-) slows hERG activation, while both F(-) and Br(-) make the channel close faster; (4) neither F(-) nor Br(-) have any effect on hERG inactivation kinetics. In conclusion, compared to Cl(-), F(-) has subtle effect on hERG activation, while Br(-) has distinct effects on certain, but not all biophysical properties of hERG channel.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/fisiología , Halógenos/farmacología , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp/métodos , Bloqueadores de los Canales de Potasio/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos/métodos , Canal de Potasio ERG1 , Citometría de Flujo/métodos , Análisis de Inyección de Flujo/métodos , Activación del Canal Iónico/efectos de los fármacos , Iones , Potenciales de la Membrana/efectos de los fármacos
3.
J Pharmacol Toxicol Methods ; 59(2): 62-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19367686

RESUMEN

INTRODUCTION: Ca(v)1.2 channels play an important role in shaping the cardiac action potential. Screening pharmaceutical compounds for Ca(v)1.2 block is very important in developing drugs without cardiac liability. Ca(v)1.2 screening has been traditionally done using fluorescence assays, but these assays have some limitations. Patch clamping is considered the gold standard for ion channel studies, but is very labor intensive. The purpose of this study was to develop a robust medium throughput Ca(v)1.2 screening assay in PatchXpress 7000A by optimizing cell isolation conditions, recording solutions and experimental parameters. Under the conditions established, structurally different standard Ca(v)1.2 antagonists and an agonist were tested. METHODS: HEK-293 cells stably transfected with hCa(v)1.2 L-type Ca channel were used. For experiments, cells were isolated using 0.05% Trypsin. Currents were recorded in the presence of 30 mM extracellular Ba2+ and low magnesium intracellular recording solution to minimize rundown. Ca(v)1.2 currents were elicited from a holding potential of -60 mV at 0.05 Hz to increase pharmacological sensitivity and minimize rundown. Test compounds were applied at increasing concentrations for 5 min followed by a brief washout. RESULTS: Averaged peak Ca(v)1.2 current amplitudes were increased from 10 pA/pF to 15 pA/pF by shortening cell incubation and trypsin exposure time from 2.5 min at 37 degrees C to 1 min at room temperature and adding 0.2 mM cAMP to the intracellular solution. Rundown was minimized from 2%/min to 0.5%/min by reducing the intracellular free Mg2+ from 2.7 mM to 0.2 mM and adding 100 nM Ca2+. Under the established conditions, we tested 8 structurally different antagonists and an agonist. The IC(50) values obtained ranked well against published values and results obtained using traditional clamp experiments performed in parallel using the expressed cell line and native myocytes. DISCUSSION: This assay can be used as a reliable pharmacological screening tool for Ca(v)1.2 block to assess compounds for cardiac liability during lead optimization.


Asunto(s)
Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Agonistas de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Evaluación Preclínica de Medicamentos/instrumentación , Nifedipino/farmacología , Técnicas de Placa-Clamp/instrumentación , Animales , Bario/metabolismo , Línea Celular , Colagenasas/farmacología , Relación Dosis-Respuesta a Droga , Electrofisiología , Cobayas , Ventrículos Cardíacos/citología , Humanos , Concentración 50 Inhibidora , Riñón/citología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Péptido Hidrolasas/farmacología , Temperatura , Factores de Tiempo , Transfección , Tripsina/farmacología
4.
Curr Top Med Chem ; 8(13): 1102-12, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18782006

RESUMEN

This review summarizes current knowledge of the cardiac rapidly activating delayed rectifier potassium current (I(Kr)), and its connection to drug-acquired QT prolongation and the associated risk of ventricular arrhythmia and fibrillation. The molecular characterization of hERG as the structural correlate of I(Kr) and the link between inherited long QT and the KCNH2 gene (hERG), have facilitated mechanistic studies of drug-acquired QT prolongation. The development of high throughput assays to evaluate drug effects on hERG has provided an avenue to determine structure activity relations (SAR) within chemical series. More than 10 years of collective data and structural considerations support the notion that hERG is an unusually promiscuous target among potassium channels, but that defining SAR within a chemical series is a viable strategy to reduce or eliminate hERG activity. Despite a critical need to minimize drug effects on hERG, one should always keep in mind that hERG is not the only structural correlate of QT prolongation, and that QT prolongation is a sub-optimal biomarker for ventricular arrhythmia and fibrillation.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/metabolismo , Evaluación Preclínica de Medicamentos , Canales de Potasio Éter-A-Go-Go/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Electrocardiografía , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Factores de Riesgo , Relación Estructura-Actividad
5.
Assay Drug Dev Technol ; 6(2): 235-41, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18471077

RESUMEN

Blockade of the human ether-a-go-go-related gene (hERG) potassium channel, with a consequent possibility of QT prolongation and increased susceptibility to a characteristic polymorphic ventricular arrhythmia, torsade de pointes, is an important cause of withdrawal of drugs from the market. In the aftermath of recent drug withdrawals, regulatory agencies now require in vitro hERG screening of all pharmaceutical compounds that are targeted for human use. To minimize the potential for failure in later-stage drug development, many pharmaceutical and biotechnology companies have begun to use automated patch clamp systems with higher throughput than conventional manual patch-clamp techniques to conduct routine functional hERG screening during drug discovery and early development. We have optimized an automated patch-clamp hERG screening method for the PatchXpress 7000A system (Molecular Devices, Sunnyvale, CA) using potassium fluoride (KF) in the internal recording solution. In this study we show that (1) the biophysical and pharmacological properties of hERG current recorded with KF are similar to those with standard potassium chloride solutions, (2) use of KF significantly improves the success rate of hERG screening using PatchXpress without compromising data quality, and (3) utilization of KF can significantly increase the throughput of hERG screening with PatchXpress.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Fluoruros , Técnicas de Placa-Clamp/métodos , Bloqueadores de los Canales de Potasio/farmacología , Compuestos de Potasio , Animales , Células CHO , Membrana Celular/efectos de los fármacos , Cricetinae , Cricetulus , Interpretación Estadística de Datos , Electrofisiología
6.
J Pharmacol Toxicol Methods ; 58(2): 72-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18508287

RESUMEN

Biological therapeutic agents (biologicals), such as monoclonal antibodies (mAbs), are increasingly important in the treatment of human disease, and many types of biologicals are in clinical development. During preclinical drug development, cardiovascular safety pharmacology studies are performed to assess cardiac safety in accord with the ICH S7A and S7B regulations that guide these studies. The question arises, however, whether or not it is appropriate to apply these guidelines, which were devised primarily to standardize small molecule drug testing, to the cardiovascular evaluation of biologicals. We examined the scientific literature and formed a consensus of scientific opinion to determine if there is a rational basis for conducting an in vitro hERG assay as part of routine preclinical cardiovascular safety testing for biologicals. We conclude that mAb therapeutics have very low potential to interact with the extracellular or intracellular (pore) domains on hERG channel and, therefore, are highly unlikely to inhibit hERG channel activity based on their targeted, specific binding properties. Furthermore, mAb are large molecules (>140,000 Da) that cannot cross plasma membranes and therefore would be unable to access and block the promiscuous inner pore of the hERG channel, in contrast with typical small molecule drugs. Consequently, we recommend that it is not appropriate to conduct an in vitro hERG assay as part of a preclinical strategy for assessing the heart rate corrected QT interval (QTc) prolongation risk of mAbs and other types of biologicals. It is more appropriate to assess QTc risk by integrating cardiovascular endpoints into repeat-dose general toxicology studies performed in an appropriate non-rodent species. These recommendations should help shape future regulatory strategy and discussions for the cardiovascular safety pharmacology testing of mAbs as well as other biologicals and provide guidance for the preclinical cardiovascular evaluation of such agents.


Asunto(s)
Productos Biológicos/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Evaluación Preclínica de Medicamentos/métodos , Humanos
7.
Assay Drug Dev Technol ; 5(5): 617-27, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17939752

RESUMEN

A cardiac safety concern for QT prolongation and potential for pro-arrhythmia exists due to inhibition of the cardiac slowly activating delayed rectifier potassium current, I(Ks). Selective inhibitors of I Ks have been shown to prolong the QT interval in animal models. On the other hand, I Ks has been considered as a target for anti-arrhythmic therapy due to certain biophysical and pharmacological properties and its expression pattern in the heart. Consequently, we have developed a method utilizing a human embryonic kidney (HEK)-293 cell line expressing KCNQ1/KCNE1 (genes that encode for the I Ks channel) as a model for screening of new compounds for I Ks activity. This study was designed (1) to establish and optimize the experimental conditions for measurement of I Ks using PatchXpress() 7000A (Molecular Devices Corporation, Sunnyvale, CA) and (2) to test the effects of I Ks inhibitors and compare the 50% inhibitory concentration (IC50) values determined with PatchXpress versus conventional patch clamp in order to validate the PatchXpress approach for higher-throughput I Ks screening. Biophysical properties of HEK/I Ks recorded with PatchXpress were similar to those recorded with conventional patch-clamp and reported in the literature. The IC50 values for I Ks block determined with PatchXpress correlated well with conventional patch-clamp values from HEK-293 cells as well as from native cardiac myocytes for the majority of compounds tested. Electrophysiological recording of I Ks expressed in HEK-293 cells with the PatchXpress is of acceptable quality for screening purposes. This approach can be utilized for functional prescreening of development compounds for I Ks inhibition either for optimizing lead anti-arrhythmic or other therapeutic candidates or to exclude compounds with the potential to prolong QT.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Evaluación Preclínica de Medicamentos/instrumentación , Canal de Potasio KCNQ1/efectos de los fármacos , Técnicas de Placa-Clamp/instrumentación , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Animales , Células CHO , Línea Celular , Química Farmacéutica , Cricetinae , Cricetulus , Interpretación Estadística de Datos , Electrofisiología , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Cobayas , Humanos , Técnicas In Vitro , Miocitos Cardíacos/efectos de los fármacos
8.
J Cardiovasc Pharmacol ; 47(2): 211-20, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16495758

RESUMEN

Flunarizine has been widely used for the management of a variety of disorders such as peripheral vascular diseases, migraine, and epilepsy. The majority of its beneficial effects have been attributed to its ability to inhibit voltage-gated Ca2+ channels in the low micromolar range, albeit non-selectively, as flunarizine has been shown to inhibit a variety of ion channels. We examined the effects of flunarizine on potassium currents through cardiac channels encoded by the human ether-a-go-go related gene (hERG) stably expressed in CHO cells. In this study, we have characterized the effect of flunarizine on biophysical properties of hERG potassium currents with standard whole-cell voltage-clamp techniques. Notably, flunarizine is a highly potent inhibitor of hERG current with an IC50 value of 5.7 nM. The effect of flunarizine on hERG potassium current is concentration and time dependent, and displays voltage dependence over the voltage range between -40 and 0 mV. At concentrations near or above the IC50, flunarizine causes a negative shift in the voltage dependence of hERG current activation and accelerates tail current deactivation. Flunarizine preferentially blocks the activated state of the channel and displays weak frequency dependence of inhibition. Flunarizine also inhibits KCNQ1/KCNE1 channel current with an IC50 of 0.76 microM.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Flunarizina/farmacología , Corazón/fisiología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Animales , Células CHO , Bloqueadores de los Canales de Calcio/química , Técnicas de Cultivo de Célula , Línea Celular , Cricetinae , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Técnicas Electrofisiológicas Cardíacas , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/fisiología , Flunarizina/química , Corazón/efectos de los fármacos , Humanos , Estructura Molecular , Técnicas de Placa-Clamp , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/fisiología
9.
J Cardiovasc Pharmacol ; 42(1): 105-12, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12827034

RESUMEN

Previous cardiac electrophysiologic studies of blockers of the slowly activating delayed rectifier (IKs) current have focused primarily on ventricular repolarization. This report summarizes an extensive in vivo cardiac electrophysiologic profile of four 1,4-benzodiazepine IKs blocker analogues (L-761334, L-763540, L-761710, and L-768673) in dogs. At 3.0 mg/kg intravenously, all four analogues elicited 14.5%-21.4% increases in ventricular refractoriness and 19.2%-22.6% increases in QTc interval. Concomitant 11.1%-13.5% increases in atrial refractoriness were noted with all four analogues. Decreases in sinus heart rate of 8.4%-17.3% were noted with all four compounds. No effects on atrial, His Purkinje, ventricular conduction or atrial and ventricular excitation were observed. One analogue, L-761710, significantly delayed atrioventricular (AV) nodal conduction (40.7+/-17.4% increase in atrial-to-His interval) and increased the AV conduction system functional refractory period 19.9+/-6.2%. The lack of effect of the other three 1,4-benzodiazepine IKs blockers on AV nodal function at dosages producing comparable effects on atrial and ventricular refractoriness suggest that the AV nodal effects of L-761710 were unrelated to IKs blockade. These findings indicate IKs plays important roles in both atrial and ventricular refractoriness as well as pacemaker function in the dog heart, suggesting potential utility for IKs blockers in the treatment of atrial and ventricular arrhythmias.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Benzodiazepinas/farmacología , Sistema de Conducción Cardíaco/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Animales , Nodo Atrioventricular/efectos de los fármacos , Benzodiazepinas/química , Fascículo Atrioventricular/efectos de los fármacos , Perros , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Inyecciones Intravenosas , Masculino , Estructura Molecular , Bloqueadores de los Canales de Potasio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA