Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Sci Rep ; 5: 17623, 2015 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-26616666

RESUMO

Vanoxerine has been in clinical trials for Parkinsonism, depression and cocaine addiction but lacked efficacy. Although a potent blocker of hERG, it produced no serious adverse events. We attributed the unexpected result to offsetting Multiple Ion Channel Effects (MICE). Vanoxerine's effects were strongly frequency-dependent and we repositioned it for treatment of atrial fibrillation and flutter. Vanoxerine terminated AF/AFL in an animal model and a dose-ranging clinical trial. Reversion to normal rhythm was associated with QT prolongation yet absent proarrhythmia markers for Torsade de Pointes (TdP). To understand the QT/TdP discordance, we used quantitative profiling and compared vanoxerine with dofetilide, a selective hERG-blocking torsadogen used for intractable AF, verapamil, a non-torsadogenic MICE comparator and bepridil, a torsadogenic MICE comparator. At clinically relevant concentrations, verapamil blocked hCav1.2 and hERG, as did vanoxerine and bepridil both of which also blocked hNav1.5. In acute experiments and simulations, dofetilide produced early after depolarizations (EADs) and arrhythmias, whereas verapamil, vanoxerine and bepridil produced no proarrhythmia markers. Of the MICE drugs only bepridil inhibited hERG trafficking following overnight exposure. The results are consistent with the emphasis on MICE of the CiPA assay. Additionally we propose that trafficking inhibition of hERG be added to CiPA.


Assuntos
Coração/efeitos dos fármacos , Canais Iônicos/metabolismo , Miocárdio/metabolismo , Piperazinas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Bepridil/farmacologia , Células CHO , Simulação por Computador , Cricetulus , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Concentração Inibidora 50 , Potenciais da Membrana/efeitos dos fármacos , Modelos Biológicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Fenetilaminas/farmacologia , Sulfonamidas/farmacologia , Verapamil/farmacologia
2.
Heart Rhythm ; 12(6): 1105-12, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25684233

RESUMO

BACKGROUND: Restoration of sinus rhythm (SR) in patients with atrial fibrillation/atrial flutter (AF/AFL) is limited principally to direct current cardioversion. The multi-ion channel blocker vanoxerine may prove an effective alternative. OBJECTIVE: The purpose of this study was to assess vanoxerine, a 1,4-dialkylpiperazine derivative, for acute conversion of recent-onset, symptomatic AF and AFL. METHODS: One hundred four subjects with symptomatic AF/AFL for <7 days were randomized sequentially to single oral doses of vanoxerine 200, 300, and 400 mg or placebo. Holter monitors were examined for conversion to SR and proarrhythmia through ≥24 hours. RESULTS: Conversion to SR was dose related: 18.2%, 44.0%, and 52.0% within 4 hours, and 59.1%, 64.0%, and 84.0% within 24 hours, for the 200-, 300-, and 400-mg groups, respectively. This was significantly higher than placebo for the 300- and 400-mg groups within 4 hours (12.5% for placebo; P = .0138 and P = .0028, respectively) and for all doses within 24 hours (31.3% for placebo; P = .0421, P = .0138, P = .0001 for 200-, 300-, and 400-mg vanoxerine groups, respectively). Although vanoxerine caused significant dose-dependent QTcF (QT correction by Fridericia) prolongation, monomorphic or polymorphic ventricular tachycardia did not occur. Adverse events were mild and self-limited, with only the highest dose having a greater frequency than placebo. CONCLUSION: Oral vanoxerine converted AF/AFL to SR at a high rate, was well tolerated, and caused no ventricular proarrhythmia.


Assuntos
Fibrilação Atrial/tratamento farmacológico , Flutter Atrial/tratamento farmacológico , Inibidores da Captação de Dopamina/administração & dosagem , Piperazinas/administração & dosagem , Administração Oral , Idoso , Método Duplo-Cego , Cardioversão Elétrica , Eletrocardiografia , Eletrocardiografia Ambulatorial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
3.
Assay Drug Dev Technol ; 12(2): 110-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24579774

RESUMO

Voltage-gated Ca2+ channels play essential roles in control of neurosecretion and muscle contraction. The pharmacological significance of Cav channels stem from their identification as the molecular targets of calcium blockers used in the treatment of cardiovascular diseases, such as hypertension, angina, and arrhythmia, and neurologic diseases, such as pain and seizure. It has been proposed that state-dependent Cav inhibitors, that is, those that preferentially bind to channels in open or inactivated states, may improve the therapeutic window over relatively state-independent Cav inhibitors. High-throughput fluorescent-based functional assays have been useful in screening chemical libraries to identify Cav inhibitors. However, hit confirmation, mechanism of action, and subtype selectivity are better suited to automated patch clamp assays that have sufficient capacity to handle the volume of compounds identified during screening, even of modest sized libraries (≤500,000 compounds), and the flexible voltage control that allows evaluation of state-dependent drug blocks. IonWorks Barracuda (IWB), the newest generation of IonWorks instruments, provides the opportunity to accelerate the Cav drug discovery studies in an automated patch clamp platform in 384-well format capable of medium throughput screening and profiling studies. We have validated hCav1.2, hCav2.1, hCav2.2, and hCav3.2 channels assays on the IWB platform (population patch clamp mode) and demonstrated that the biophysical characteristics of the channels (activation, inactivation, and steady-state inactivation) obtained with the IWB system are consistent with known subtype-specific characteristics. Using standard reference compounds (nifedipine, BAY K8644, verapamil, mibefradil, and pimozide), we demonstrated subtype-selective and state- and use-dependent characteristics of drug-channel interactions. Here we describe the design and validation of novel robust high-throughput Cav channel assays on the IWB platform. The assays can be used to screen focused compound libraries for state-dependent Cav channel antagonists, to prioritize compounds for potency or to counterscreen for Cav subtype selectivity.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Avaliação de Medicamentos/métodos , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia
4.
Sci Rep ; 3: 2100, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23812503

RESUMO

Drug-induced block of the cardiac hERG (human Ether-à-go-go-Related Gene) potassium channel delays cardiac repolarization and increases the risk of Torsade de Pointes (TdP), a potentially lethal arrhythmia. A positive hERG assay has been embraced by regulators as a non-clinical predictor of TdP despite a discordance of about 30%. To test whether assaying concomitant block of multiple ion channels (Multiple Ion Channel Effects or MICE) improves predictivity we measured the concentration-responses of hERG, Nav1.5 and Cav1.2 currents for 32 torsadogenic and 23 non-torsadogenic drugs from multiple classes. We used automated gigaseal patch clamp instruments to provide higher throughput along with accuracy and reproducibility. Logistic regression models using the MICE assay showed a significant reduction in false positives (Type 1 errors) and false negatives (Type 2 errors) when compared to the hERG assay. The best MICE model only required a comparison of the blocking potencies between hERG and Cav1.2.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Modelos Teóricos , Torsades de Pointes/fisiopatologia , Canal de Potássio ERG1 , Humanos , Técnicas de Patch-Clamp , Valor Preditivo dos Testes , Torsades de Pointes/diagnóstico
5.
Toxicol Appl Pharmacol ; 272(1): 245-55, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23707608

RESUMO

Tyrosine kinase inhibitors (TKi) have greatly improved the treatment and prognosis of multiple cancer types. However, unexpected cardiotoxicity has arisen in a subset of patients treated with these agents that was not wholly predicted by pre-clinical testing, which centers around animal toxicity studies and inhibition of the human Ether-à-go-go-Related Gene (hERG) channel. Therefore, we sought to determine whether a multi-parameter test panel assessing the effect of drug treatment on cellular, molecular, and electrophysiological endpoints could accurately predict cardiotoxicity. We examined how 4 FDA-approved TKi agents impacted cell viability, apoptosis, reactive oxygen species (ROS) generation, metabolic status, impedance, and ion channel function in human cardiomyocytes. The 3 drugs clinically associated with severe cardiac adverse events (crizotinib, sunitinib, nilotinib) all proved to be cardiotoxic in our in vitro tests while the relatively cardiac-safe drug erlotinib showed only minor changes in cardiac cell health. Crizotinib, an ALK/MET inhibitor, led to increased ROS production, caspase activation, cholesterol accumulation, disruption in cardiac cell beat rate, and blockage of ion channels. The multi-targeted TKi sunitinib showed decreased cardiomyocyte viability, AMPK inhibition, increased lipid accumulation, disrupted beat pattern, and hERG block. Nilotinib, a second generation Bcr-Abl inhibitor, led to increased ROS generation, caspase activation, hERG block, and an arrhythmic beat pattern. Thus, each drug showed a unique toxicity profile that may reflect the multiple mechanisms leading to cardiotoxicity. This study demonstrates that a multi-parameter approach can provide a robust characterization of drug-induced cardiomyocyte damage that can be leveraged to improve drug safety during early phase development.


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteínas Quinases/toxicidade , Proteínas Tirosina Quinases/antagonistas & inibidores , Caspase 3/metabolismo , Caspase 7/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colesterol/metabolismo , Crizotinibe , Canal de Potássio ERG1 , Ativação Enzimática/efeitos dos fármacos , Cloridrato de Erlotinib , Canais de Potássio Éter-A-Go-Go/biossíntese , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Indóis/toxicidade , Canais Iônicos/efeitos dos fármacos , Lipídeos/biossíntese , Miócitos Cardíacos/ultraestrutura , Técnicas de Patch-Clamp , Células-Tronco Pluripotentes/efeitos dos fármacos , Pirazóis/toxicidade , Piridinas/toxicidade , Pirimidinas/toxicidade , Pirróis/toxicidade , Quinazolinas/toxicidade , RNA/biossíntese , RNA/isolamento & purificação , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Sunitinibe
6.
J Pharmacol Toxicol Methods ; 68(1): 13-22, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23538024

RESUMO

The human ether-a-go-go-related gene (hERG, Kv11.1) K(+) channel plays an important role in cardiac repolarization. Following its cloning and expression it was established that inhibition of this channel was the molecular mechanism for many non-antiarrhythmic drugs that produce torsades de pointes associated with QT prolongation. Therefore the study of in vitro drug-hERG interactions has become an important part of modern safety pharmacology. Manual and automated patch clamp electrophysiology, in silico modeling, and hERG trafficking assays have been developed to aid in this study. The correlation between in vitro hERG IC50, drug exposure, QT prolongation in the thorough QT clinical trial and risk of TdP has greatly reduced drug withdrawals due to TdP. However a significant association with Type 1 errors in particular remains and may have a negative impact on drug development. Combining hERG data with other non-clinical and clinical markers of proarrhythmia will increase the specificity and sensitivity of cardiac risk assessment. hERG will continue to play an important role in drug development and safety pharmacology in the future.


Assuntos
Desenho de Fármacos , Canais de Potássio Éter-A-Go-Go/metabolismo , Síndrome do QT Longo/induzido quimicamente , Animais , Simulação por Computador , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Canal de Potássio ERG1 , Humanos , Síndrome do QT Longo/prevenção & controle , Técnicas de Patch-Clamp , Medição de Risco/métodos , Sensibilidade e Especificidade , Torsades de Pointes/induzido quimicamente , Torsades de Pointes/prevenção & controle
7.
J Cardiovasc Electrophysiol ; 22(11): 1266-73, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21615815

RESUMO

BACKGROUND: Vanoxerine is a promising, new, investigational antiarrhythmic drug. The purpose of this study was to test the hypothesis that oral dosing of vanoxerine would first terminate induced atrial flutter (AFL) and atrial fibrillation (AF), and then prevent their reinduction. METHODS: In 5 dogs with sterile pericarditis, on the fourth day after creating the pericarditis, we performed electrophysiologic (EP) studies at baseline, measuring atrial excitability, refractoriness (AERP), and conduction time (CT) when pacing from the right atrial appendage, Bachmann's bundle (BB), and the posteroinferior left atrium at cycle lengths (CLs) of 400, 300, and 200 ms. Then, after induction of AFL or AF, all dogs received hourly oral doses of vanoxerine: 90 mg, followed by 180 mg and 270 mg. Blood was obtained to determine plasma vanoxerine concentrations at baseline, every 30 minutes, when neither AFL nor AF were inducible, and, finally, 1 hour after the 270 mg dose. Then we repeated the baseline EP studies. RESULTS: Four dogs had inducible, sustained AFL, and 1 dog only had induced, nonsustained AF. In 4 AFL episodes, oral vanoxerine terminated the AFL and then rendered it noninducible after an average of 111 minutes (range 75-180 minutes) after the first dose was administered. The mean vanoxerine plasma level at the point of noninducibility was 84 ng/mL, with a narrow range of 76-99 ng/mL. In the dog with induced, nonsustained AF, it was no longer inducible at a drug level of 75 ng/mL. Vanoxerine did not significantly (1) prolong the AERP except at BB, and then only at the faster pacing CLs; (2) change atrial excitability thresholds; (3) prolong atrial conduction time, the PR interval, the QRS complex or the QT interval. CONCLUSIONS: Orally administered vanoxerine effectively terminated AFL and rendered it noninducible. It also suppressed inducibility of nonsustained AF. These effects occurred at consistent plasma drug levels. Vanoxerine's insignificant or minimal effects on measured electrophysiologic parameters are consistent with little proarrhythmic risk.


Assuntos
Antiarrítmicos/administração & dosagem , Fibrilação Atrial/tratamento farmacológico , Flutter Atrial/tratamento farmacológico , Piperazinas/administração & dosagem , Administração Oral , Animais , Antiarrítmicos/sangue , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/etiologia , Fibrilação Atrial/fisiopatologia , Flutter Atrial/diagnóstico , Flutter Atrial/etiologia , Flutter Atrial/fisiopatologia , Estimulação Cardíaca Artificial , Modelos Animais de Doenças , Cães , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Pericardite/complicações , Piperazinas/sangue , Prevenção Secundária , Fatores de Tempo
8.
J Cardiovasc Pharmacol ; 56(4): 420-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20930594

RESUMO

Methadone, a synthetic opioid for treatment of chronic pain and withdrawal from opioid dependence, has been linked to QT prolongation, potentially fatal torsades de pointes, and sudden cardiac death. Concomitant use of diazepam or other benzodiazepines in methadone maintenance treatment can increase the risk of sudden death. Therefore, we determined the effects of methadone and diazepam singly and in combination on cardiac action potentials (APs) and on the major ion channels responsible for cardiac repolarization. Using patch clamp recording in human stem cell-derived cardiomyocytes and stably transfected mammalian cells, we found that methadone produced concentration-dependent AP prolongation and ion channel block at low micromolar concentrations: hERG (IC50 = 1.7 µM), hNav1.5 (11.2 µM tonic block; 5.5 µM phasic block), and hCav1.2 (26.7 µM tonic block; 7.7 µM phasic block). Methadone was less potent in hKv4.3/hKChIP2.2 (IC50 = 39.0 µM) and hKvLQT1/hminK (53.3 µM). In contrast, diazepam blocked channels only at much higher concentrations and had no effect on AP duration at 1 µM. However, coadministration of 1-µM diazepam with methadone caused a statistically significant increase in AP duration and a 4-fold attenuation of hNav1.5 block (IC50 values were 44.2 µM and 26.6 µM, respectively, for tonic and phasic block), with no significant effect on methadone-induced block of hERG, hCav1.2, hKv4.3/hKChIP2.2, and hKvLQT1/hminK channels. Thus, although diazepam alone does not prolong the QT interval, the relief of methadone-induced Na channel block may leave hERG K channel block uncompensated, thereby increasing cardiac risk.


Assuntos
Diazepam/efeitos adversos , Hipnóticos e Sedativos/efeitos adversos , Canais Iônicos/fisiologia , Metadona/efeitos adversos , Entorpecentes/efeitos adversos , Potenciais de Ação/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Células Cultivadas , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Humanos , Hipnóticos e Sedativos/farmacologia , Canais Iônicos/antagonistas & inibidores , Canal de Potássio KCNQ1/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Metadona/farmacologia , Proteínas Musculares/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Entorpecentes/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Canais de Sódio
9.
J Pharmacol Toxicol Methods ; 61(3): 277-86, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20153443

RESUMO

INTRODUCTION: The cardiac action potential (CAP) of stem cell-derived human cardiomyocytes (SC-hCMs) is potentially the most powerful preclinical biomarker for cardiac safety and efficacy in humans. Our experiments tested this hypothesis by examining the CAP and relevant pharmacology of these cells. METHODS: The electrophysiological and pharmacological profiles of SC-hCMs were compared to rabbit and canine Purkinje fibers (PFs). Ventricular SC-hCMs provided the dominant electrophysiological phenotype (approximately 82%) in a population of ventricular, atrial and nodal cardiomyocytes (CMs). The effects of reference compounds were measured in SC-hCMs using perforated patch, current clamp recording. Selective inhibitors of I(Kr), I(Ks), I(Ca,L), and I(Na), and norepinephrine (NE), were tested on SC-hCM action potentials (APs). RESULTS: AP prolongation was observed upon exposure to hERG channel blockers (terfenadine, quinidine, cisapride, sotalol, E-4031 and verapamil), with significantly shorter latencies than in PF assays. For the torsadogenic compounds, terfenadine and quinidine, SC-hCM AP prolongation occurred at significantly lower concentrations than in canine or rabbit PF APs. Moreover, the I(Ks) blocker chromanol 293B prolonged APs from SC-hCMs, whereas both rabbit and canine PF assays are insensitive to I(Ks) blockers in the absence of adrenergic preconditioning. Early afterdepolarizations (EADs) were induced by 100 nM E-4031 and 100 nM cisapride in the SC-hCM assay, but not in the canine or rabbit PF assay. Selective inhibition of I(Na) and I(Ca,L) slowed V(max) and shortened AP duration, respectively. NE prolonged the AP duration of SC-hCMs. DISCUSSION: The CAP of SC-hCMs has been validated as a powerful preclinical biomarker for cardiac safety and efficacy. In addition to its human nature, the SC-hCM AP assay removes diffusion delays, reduces test compound consumption, demonstrates an overall pharmacological sensitivity that is greater than conventional rabbit or canine PF assays, and accurately predicts cardiac risk of known torsadogenic compounds.


Assuntos
Potenciais de Ação/fisiologia , Células-Tronco Embrionárias/fisiologia , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Cães , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Coelhos
10.
J Cardiovasc Electrophysiol ; 21(3): 301-10, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19817928

RESUMO

INTRODUCTION: There remains an unmet need for safe and effective antiarrhythmic drugs, especially for the treatment of atrial fibrillation. Vanoxerine is a drug that is free of adverse cardiac events in normal volunteers, yet is a potent blocker of the hERG (hK(v)11.1) cardiac potassium channel. Consequently,we hypothesized that vanoxerine might also be a potent blocker of cardiac calcium (Ca) and sodium (Na) currents, and would not affect transmural dispersion of repolarization. METHODS: The whole cell patch clamp technique was used to measure currents from cloned ion channels overexpressed in stable cell lines and single ventricular myocytes. We measured intracellular action potentials from canine ventricular wedges and Purkinje fibers using sharp microelectrode technique. RESULTS: We found that vanoxerine was a potent hK(v)11.1 blocker, and at submicromolar concentrations, it blocked Ca and Na currents in a strongly frequency-dependent manner. In the canine ventricular wedge preparation vanoxerine did not significantly affect transmural action potential waveforms, QT interval or transmural dispersion of repolarization. CONCLUSIONS: Vanoxerine (1) is a potent blocker of cardiac hERG, Na and Ca channels; (2) block is strongly frequency-dependent especially for Na and Ca channels; and (3) transmural dispersion of ventricular repolarization is unaffected. The multichannel block and repolarization uniformity resemble the effects of amiodarone, the exemplar atrial fibrillation drug. Vanoxerine is a completely different chemical and has none of amiodarone's toxic effects. Vanoxerine has characteristics of a potentially effective and safe antiarrhythmic.


Assuntos
Rim/efeitos dos fármacos , Rim/metabolismo , Piperazinas/administração & dosagem , Piperazinas/metabolismo , Animais , Antiarrítmicos/administração & dosagem , Antiarrítmicos/farmacocinética , Linhagem Celular , Cães , Avaliação Pré-Clínica de Medicamentos , Humanos
11.
J Cardiovasc Electrophysiol ; 21(3): 311-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19817929

RESUMO

BACKGROUND: Vanoxerine produces potent block of cardiac hERG, sodium, and L-type calcium channels. Block is strongly frequency dependent, is unassociated with transmural dispersion of repolarization, and occurs at concentrations safe in humans. Therefore, we proposed that vanoxerine might be antiarrhythmic. In these studies, we tested the hypothesis that vanoxerine would terminate induced atrial fibrillation (AF) and atrial flutter (AFL) in dogs with sterile pericarditis (SP). METHODS AND RESULTS: In 9 SP dogs, 11 episodes each of sustained (>10 minutes) AF and AFL were induced. Electrophysiological studies were performed before and after infusion of vanoxerine, which effectively terminated AF and AFL in 19 of 22 episodes. Simultaneous multisite mapping during 3 AF and 3 AFL episodes demonstrated that termination of each arrhythmia occurred with termination of the driver (a reentrant circuit) following an increase in tachycardia CL. Except for conduction in an area of slow conduction in the driver's reentrant circuit, vanoxerine did not significantly affect intraatrial or atrioventricular conduction time, QRS duration, or QT/QTc intervals. Ventricular refractoriness prolonged minimally during ventricular pacing at 400 and 333 ms (176 +/- 16 ms to 182 +/- 16 ms; 173 +/- 11 ms to 178 +/- 18 ms, respectively). Vanoxerine minimally increased (mean 0.7 mA) atrial stimulus threshold for capture. CONCLUSIONS: Vanoxerine effectively terminated induced, sustained AF and AFL in the canine SP model, and produced insignificant or minimal changes in refractoriness, conduction time, or stimulus threshold, consistent with little proarrhythmic risk.


Assuntos
Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/fisiopatologia , Flutter Atrial/tratamento farmacológico , Flutter Atrial/fisiopatologia , Eletrocardiografia/efeitos dos fármacos , Frequência Cardíaca/efeitos da radiação , Piperazinas/administração & dosagem , Animais , Antiasmáticos/administração & dosagem , Fibrilação Atrial/diagnóstico , Flutter Atrial/diagnóstico , Cães , Resultado do Tratamento
12.
J Pharmacol Toxicol Methods ; 59(1): 29-34, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19028590

RESUMO

INTRODUCTION: Drug-induced prolongation of the electrocardiographic QT interval (long QT syndrome) has been associated with increased risk of a serious ventricular arrhythmia, torsade de pointes. Inhibition of hERG, a cardiac potassium channel that controls action potential repolarization, is the most common cause of QT prolongation by non-cardiac drugs. The ICH S7B describes preclinical safety testing required for new drugs, including the determination of the hERG IC(50). Actual and target concentrations may differ due to solubility, stability, or loss of compound. Significant differences will invalidate quantitative concentration-response curves which may be critical to interpretation of drug safety. To examine the frequency and significance of these differences, we conducted an analysis of studies where both the electrophysiology and the dose solution analysis were conducted in-house. We have investigated the actual concentrations of test article in vehicle solution as compared to the target concentrations in an attempt to determine the reasons behind differences between the two values. METHODS: Studies that involved both electrophysiology and dose solution analysis performed at ChanTest Corporation were evaluated. The effects of stability, solubility and loss through the perfusion apparatus on actual dosing concentrations were investigated. RESULTS: There was a large range in the loss of the test article attributed to the perfusion apparatus (range from 0 to 74% loss). For 12 of the 22 studies evaluated, the IC(50) was>2-fold more potent when using actual values as determined by HPLC versus the target concentrations. Twenty-two percent of the test articles were not stable 24 h after room temperature storage; 16% after 24 h frozen conditions. DISCUSSION: The best practices when considering dose solution concentration verification of test article solutions are to: determine the solubility of the compound in a physiological buffer, analyze samples collected from the perfusion chamber, and analyze samples the same day as sample collection (e.g., same day as hERG assay).


Assuntos
Canais de Potássio Éter-A-Go-Go/análise , Perfusão/instrumentação , Preparações Farmacêuticas/análise , Soluções Tampão , Dimetil Sulfóxido/química , Eletrofisiologia/métodos , Etanol/química , Estudos de Avaliação como Assunto , Humanos , Preparações Farmacêuticas/química , Veículos Farmacêuticos/análise , Veículos Farmacêuticos/química , Farmacologia/métodos , Padrões de Referência , Soluções/análise , Solventes/química , Estudos de Validação como Assunto
13.
Eur Biophys J ; 38(3): 273-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18677468

RESUMO

Development of a large, representative library of ion channel-expressing cell lines is described. Validation on a full range of automated patch clamp and fluorescence illumination platforms is ongoing. Library "books" can be mixed and matched into channel panels according to tissue, therapeutic area and ion channel family. Unexpected results using a cardiac channel panel show that this panel may serve as a biomarker for cardiac risk assessment.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Canais Iônicos/fisiologia , Farmacologia , Animais , Automação , Linhagem Celular , Cricetinae , Cricetulus , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/biossíntese , Canais de Potássio Éter-A-Go-Go/fisiologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/biossíntese , Canais Iônicos/genética , Miocárdio/metabolismo , Técnicas de Patch-Clamp
14.
Assay Drug Dev Technol ; 6(6): 765-80, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19090691

RESUMO

Ion channels represent the third largest class of targets in drug discovery after G-protein coupled receptors and kinases. In spite of this ranking, ion channels continue to be under exploited as drug targets compared with the other two groups for several reasons. First, with 400 ion channel genes and an even greater number of functional channels due to mixing and matching of individual subunits, a systematic collection of ion channel-expressing cell lines for drug discovery and safety screening has not been available. Second, the lack of high-throughput functional assays for ion channels has limited their use as drug targets. Now that automated electrophysiology has come of age and provided the technology to assay ion channels at medium to high throughput, we have addressed the need for a library of ion channel cell lines by constructing the Ion Channel Panel (ChanTest Corp., Cleveland, OH). From 400 ion channel genes, a collection of 82 of the most relevant human ion channels for drug discovery, safety, and human disease has been assembled.Each channel has been stably overexpressed in human embryonic kidney 293 or Chinese hamster ovary cells. Cell lines have been selected and validated on automated electrophysiology systems to facilitate cost-effective screening for safe and selective compounds at earlier stages in the drug development process. The screening and validation processes as well as the relative advantages of different screening platforms are discussed.


Assuntos
Canais Iônicos/química , Animais , Astemizol/farmacologia , Astemizol/normas , Automação , Células CHO , Linhagem Celular , Clonagem de Organismos , Cricetinae , Cricetulus , DNA Complementar/genética , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos/economia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Corantes Fluorescentes/metabolismo , Humanos , Concentração Inibidora 50 , Canais Iônicos/genética , Pimozida/farmacologia , Pimozida/normas , Terfenadina/farmacologia , Terfenadina/normas
15.
J Pharmacol Exp Ther ; 324(2): 427-33, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17986649

RESUMO

The United States Food and Drug Administration (FDA) uses alfuzosin as an example of a drug having QT risk in humans that was not detected in nonclinical studies. FDA approval required a thorough clinical QT study (TCQS) that was weakly positive at high doses. The FDA has used the clinical/nonclinical discordance as a basis for mandatory TCQS, and this requirement has serious consequences for drug development. For this reason, we re-examined whether nonclinical signals of QT risk for alfuzosin were truly absent. Alfuzosin significantly prolonged action potential duration (APD)(60) in rabbit Purkinje fibers (p < 0.05) and QT in isolated rabbit hearts (p < 0.05) at the clinically relevant concentration of 300 nM. In man, the QT interval corrected with Fridericia's formula increased 7.7 ms, which exceeds the 5.0-ms threshold for a positive TCQS. Effects on hK(v)11.1, hK(v)4.3, and hK(v)7.1/hKCNE1 potassium currents and calcium current were not involved. At 300 nM, approximately 30x C(max), alfuzosin significantly increased whole-cell peak sodium (hNa(v)1.5) current (p < 0.05), increased the probability of late hNa(v)1.5 single-channel openings, and significantly shortened the slow time constant for recovery from inactivation. Alfuzosin also increased hNa(v)1.5 burst duration and number of openings per burst between 2- and 3-fold. Alfuzosin is a rare example of a non-antiarrhythmic drug that delays cardiac repolarization not by blocking hK(v)11.1 potassium current, but by increasing sodium current. Nonclinical studies clearly show that alfuzosin increases plateau potential and prolongs APD and QT, consistent with QT prolongation in man. The results challenge the FDA grounds for the absolute primacy of TCQS based on the claim of a false-negative, nonclinical study on alfuzosin.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Quinazolinas/farmacologia , Potenciais de Ação/fisiologia , Animais , Linhagem Celular , Feminino , Cobaias , Humanos , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/fisiopatologia , Masculino , Miócitos Cardíacos/fisiologia , Ramos Subendocárdicos/efeitos dos fármacos , Ramos Subendocárdicos/fisiologia , Quinazolinas/uso terapêutico , Coelhos
16.
Assay Drug Dev Technol ; 6(6): 737-42, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19231939

RESUMO

Dr. Arthur M. "Buzz" Brown is the founder and CEO of ChanTest Corporation, an ion channel company specializing in drug discovery and safety services. He is Adjunct Professor of Physiology and Biophysics, School of Medicine, Case Western Reserve University. Dr. Brown has more than 30 years of experience in ion channel structure-function relationships and their associations with human health. He established world-leading ion channel departments at University of Texas Medical Branch, Baylor College of Medicine, and Case Western Reserve University. His lab first applied liquid ion exchanger ion-selective microelectrodes to single cells, introduced the concept of membrane delimited action of G proteins on ion channels, identified the ion channel conduction pathway or pore of voltage-gated channels and inwardly rectifying potassium channels, showed that the human ether-à-go-go-related gene potassium channel was the molecular target for lethal arrhythmias associated with noncardiac drugs, and established that noncardiac drugs could also produce lethal arrhythmias by inhibiting ion channel trafficking. Dr. Brown holds eight patents on ion channel methodology and application of ion channel pharmacology to therapeutics.


Assuntos
Descoberta de Drogas/organização & administração , Descoberta de Drogas/tendências , Pesquisa Biomédica , Ensaios Clínicos Fase II como Assunto , Descoberta de Drogas/métodos , Indústria Farmacêutica , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Canais Iônicos/classificação , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/uso terapêutico , Masculino , Patentes como Assunto , Ensino
17.
Novartis Found Symp ; 266: 57-69; discussion 70-4, 95-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16050262

RESUMO

Mutations in the cardiac potassium channel hERG/IKr cause inherited long QT syndrome with increased susceptibility to ventricular arrhythmias. Several mutations in hERG produce trafficking-deficient channels that are retained in the endoplasmic reticulum (ER). Surface expression of certain mutations (i.e. hERG G601S) can be restored by specific channel blockers. Although hERG currents have been studied extensively, little is known about proteins in the processing pathway. Using biochemical and electrophysiological assays we show that the cytosolic chaperones Hsp70 and Hsp90 interact transiently with wild-type hERG. Inhibition of Hsp90 prevents maturation and reduces hERG/IKr currents. Trafficking-deficient mutants remain tightly associated with chaperones in the ER until trafficking is restored, e.g. by channel blockers. hERG/chaperone complexes represent novel targets for therapeutic compounds with cardiac liability such as arsenic, which is used in the treatment of leukaemias. Arsenic interferes with the formation of hERG/chaperone complexes and inhibits hERG maturation causing ECG abnormalities. We conclude that Hsp9O and Hsp70 are crucial for productive folding of wild-type hERG. Therapeutic compounds that inhibit chaperone function produce a novel form of acquired long QT syndrome not by direct channel block but by reduced surface expression due to an acquired trafficking defect of hERG.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP70/fisiologia , Proteínas de Choque Térmico HSP90/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Potenciais de Ação , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Humanos , Síndrome do QT Longo , Transporte Proteico
18.
Novartis Found Symp ; 266: 118-31; discussion 131-5, 155-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16050265

RESUMO

Non-cardiac drugs may prolong action potential duration (APD) and QT leading to Torsade de Pointes (TdP) and sudden cardiac death. TdP is rare and QT is used as a surrogate marker in the clinic. For non-cardiac drugs, APD/QT liability is always associated with a reduction in hERG current produced by either direct channel block or inhibition of trafficking. hERG and APD liabilities correlate better when APDs are measured in rabbit versus canine Purkinje fibres. hERG and APD/QT liabilities may be dissociated when hERG block is offset by block of calcium or sodium currents. hERG liability may be placed in context by calculating a safety margin (SM) from the IC50 for inhibition of hERG current measured by patch clamp divided by the effective therapeutic plasma concentration of the drug. The SM is uncertain because literature values for IC50 may vary by 50-fold and small differences in plasma protein binding have large effects. With quality control, the IC50 95% confidence limits vary less than twofold. Ideally, hERG liability should be determined during lead optimization. Patch damp has insufficient throughput for this purpose. A novel high-throughput screen has been developed to detect drugs that block hERG directly and/or inhibit hERG trafficking.


Assuntos
Antiarrítmicos/efeitos adversos , Morte Súbita Cardíaca/etiologia , Síndrome do QT Longo/etiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Torsades de Pointes/etiologia , Potenciais de Ação , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Humanos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo
19.
J Pharmacol Toxicol Methods ; 52(1): 136-45, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15950494

RESUMO

INTRODUCTION: Direct block of I(Kr) by non-antiarrhythmic drugs (NARDs) is a major cause of QT prolongation and torsades de pointes (TdP), and has made the hERG potassium channel a major target of drug safety programs in cardiotoxicity. Block of hERG currents is not the only way that drugs can adversely impact the repolarizing current I(Kr), however. We have shown recently that two drugs in clinical use do not block hERG but produce long QT syndrome (LQTS) and TdP by inhibiting trafficking of hERG to the cell surface. To address the need for an inexpensive, rapid, and comprehensive assay to predict both types of hERG risk early in the drug development process, we have developed a novel antibody-based chemiluminescent assay called HERG-Lite. METHODS: HERG-Lite monitors the expression of hERG at the cell surface in two different stable mammalian cell lines. One cell line acts as a biosensor for drugs that inhibit hERG trafficking, while the other predicts hERG blockers based on their ability to act as pharmacological chaperones. In this study, we have validated the HERG-Lite assay using a panel of 100 drugs: 50 hERG blockers and 50 nonblockers. RESULTS: HERG-Lite correctly predicted hERG risk for all 100 test compounds with no false positives or negatives. All 50 hERG blockers were detected as drugs with hERG risk in the HERG-Lite assay, and fell into two classes: B (for blocker) and C (for complex; block and trafficking inhibition). DISCUSSION: HERG-Lite is the most comprehensive assay available for predicting drug-induced hERG risk. It accurately predicts both channel blockers and trafficking inhibitors in a rapid, cost-effective manner and is a valuable non-clinical assay for drug safety testing.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Síndrome do QT Longo/induzido quimicamente , Bloqueadores dos Canais de Potássio/efeitos adversos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Torsades de Pointes/induzido quimicamente , Linhagem Celular , Humanos , Medições Luminescentes , Preparações Farmacêuticas/classificação , Bloqueadores dos Canais de Potássio/classificação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/imunologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Valor Preditivo dos Testes
20.
J Pharmacol Exp Ther ; 312(1): 316-23, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15340016

RESUMO

The diamidine pentamidine is used to treat leishmaniasis, trypanosomiasis, and Pneumocystis carinii pneumonia. Treatment may be accompanied by prolongation of the QT interval of the electrocardiogram and torsades de pointes tachycardias. Up to now, it has been thought that therapeutic compounds causing QT prolongation are associated with direct block of the cardiac potassium channel human ether a-go-go-related gene (hERG), which encodes the alpha subunit of cardiac I(Kr) currents. We show that pentamidine has no acute effects on currents produced by hERG, KvLQT1/mink, Kv4.3, or SCNA5. Cardiac calcium currents and the guinea pig cardiac action potential were also not affected. After overnight exposure, however, pentamidine reduced hERG currents and inhibited trafficking and maturation of hERG with IC(50) values of 5 to 8 microM similar to therapeutic concentrations. Surface expression determined in a chemiluminescence assay was reduced on exposure to 10, 30, and 100 microM pentamidine by about 30, 40, and 70%, respectively. These effects were specific for hERG since expression of hKv1.5, KvLQT1/minK, and Kv4.3 was not altered. In isolated guinea pig ventricular myocytes, 10 microM pentamidine prolonged action potential duration APD(90) from 374.3 +/- 57.1 to 893.9 +/- 86.2 ms on overnight incubation. I(Kr) tail current density was reduced from 0.61 +/- 0.09 to 0.39 +/- 0.04 pA/pF. We conclude that pentamidine prolongs the cardiac action potential by block of hERG trafficking and reduction of the number of functional hERG channels at the cell surface. We propose that pentamidine, like arsenic trioxide, produces QT prolongation and torsades de pointes in patients by inhibition of hERG trafficking.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Síndrome do QT Longo , Miócitos Cardíacos/efeitos dos fármacos , Pentamidina/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Antifúngicos/farmacologia , Transporte Biológico/efeitos dos fármacos , Células CHO , Proteínas de Transporte de Cátions/antagonistas & inibidores , Células Cultivadas , Cricetinae , Eletrofisiologia , Canais de Potássio Éter-A-Go-Go , Cobaias , Humanos , Miócitos Cardíacos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...