Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Toxicol ; 43(4): 357-367, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38477622

RESUMEN

In silico modeling offers an opportunity to supplement and accelerate cardiac safety testing. With in silico modeling, computational simulation methods are used to predict electrophysiological interactions and pharmacological effects of novel drugs on critical physiological processes. The O'Hara-Rudy's model was developed to predict the response to different ion channel inhibition levels on cardiac action potential duration (APD) which is known to directly correlate with the QT interval. APD data at 30% 60% and 90% inhibition were derived from the model to delineate possible ventricular arrhythmia scenarios and the marginal contribution of each ion channel to the model. Action potential values were calculated for epicardial, myocardial, and endocardial cells, with action potential curve modeling. This study assessed cardiac ion channel inhibition data combinations to consider when undertaking in silico modeling of proarrhythmic effects as stipulated in the Comprehensive in Vitro Proarrhythmia Assay (CiPA). As expected, our data highlight the importance of the delayed rectifier potassium channel (IKr) as the most impactful channel for APD prolongation. The impact of the transient outward potassium channel (Ito) inhibition on APD was minimal while the inward rectifier (IK1) and slow component of the delayed rectifier potassium channel (IKs) also had limited APD effects. In contrast, the contribution of fast sodium channel (INa) and/or L-type calcium channel (ICa) inhibition resulted in substantial APD alterations supporting the pharmacological relevance of in silico modeling using input from a limited number of cardiac ion channels including IKr, INa, and ICa, at least at an early stage of drug development.


Asunto(s)
Potenciales de Acción , Simulación por Computador , Canales Iónicos , Miocitos Cardíacos , Potenciales de Acción/efectos de los fármacos , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Canales Iónicos/efectos de los fármacos , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología
2.
Biomedicines ; 10(10)2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36289616

RESUMEN

The importance of magnesium (Mg2+), a micronutrient implicated in maintaining and establishing a normal heart rhythm, is still controversial. It is known that magnesium is the cofactor of 600 and the activator of another 200 enzymatic reactions in the human organism. Hypomagnesemia can be linked to many factors, causing disturbances in energy metabolism, ion channel exchanges, action potential alteration and myocardial cell instability, all mostly leading to ventricular arrhythmia. This review article focuses on identifying evidence-based implications of Mg2+ in cardiac arrhythmias. The main identified benefits of magnesemia correction are linked to controlling ventricular response in atrial fibrillation, decreasing the recurrence of ventricular ectopies and stopping episodes of the particular form of ventricular arrhythmia called torsade de pointes. Magnesium has also been described to have beneficial effects on the incidence of polymorphic ventricular tachycardia and supraventricular tachycardia. The implication of hypomagnesemia in the genesis of atrial fibrillation is well established; however, even if magnesium supplementation for rhythm control, cardioversion facility or cardioversion success/recurrence of AF after cardiac surgery and rate control during AF showed some benefit, it remains controversial. Although small randomised clinical trials showed a reduction in mortality when magnesium was administered to patients with acute myocardial infarction, the large randomised clinical trials failed to show any benefit of the administration of intravenous magnesium over placebo.

3.
Cardiovasc J Afr ; 33(5): 277-281, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36162825

RESUMEN

Sheehan's syndrome is a type of hypopituitarism caused by massive uterine bleeding and hypovolaemic shock after or during delivery. Heart involvement has been documented sporadically among the various clinical manifestations of Sheehan's syndrome but life-threatening arrhythmias are infrequent. Here, we report on two rare cases of ventricular tachycardia caused by Sheehan's syndrome. Both female patients were diagnosed with Sheehan's syndrome 30 years previously, due to massive postpartum bleeding. Both of them terminated hormone replacement therapy recently. Both patients presented with torsade de pointes. The electrocardiogram showed prolonged QT interval. In addition to potassium supplementation and anti-arrhythmia therapy, steroids and thyroid hormone replacement therapy were employed, QT-interval prolongation and T-wave inversion were normalised, and implantable cardioverter defibrillator implantation was avoided. One of the patients was recovering well at the one-year follow up and the other patient was in a coma at the time of this report. We also review the literature for cases of Sheehan's syndrome presenting with ventricular tachycardia.


Asunto(s)
Hipopituitarismo , Hemorragia Posparto , Taquicardia Ventricular , Humanos , Femenino , Hipopituitarismo/complicaciones , Hipopituitarismo/diagnóstico , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/etiología , Taquicardia Ventricular/terapia , Electrocardiografía , Periodo Posparto
4.
Front Cardiovasc Med ; 9: 903354, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711361

RESUMEN

Torsades de Pointes (TdP) occurred in a 68-year-old female with epidermal growth factor receptor (EGFR) mutant lung cancer administered osimertinib, the third-generation EGFR tyrosine kinase inhibitor (TKI). Electrocardiogram (ECG) recorded at Tdp showed QT prolongation (QTc = 515 ms), to which a Traditional Chinese Medicine (TCM) named "Litsea Cubeba" may have contributed. After discontinuation of osimertinib and Litsea Cubeba, magnesium supplementation, potassium supplementation, lidocaine infusion, and the pacemaker frequency adjustment, Tdp terminated. However, QT prolongation sustained at discharge (QTc = 528 ms), partly because of the emergency use of amiodarone. Osimertinib may prolong the QT interval leading to TdP, especially when multiple risk factors to lengthen QT interval are incidentally overlapped. Thus, regular monitoring of ECG and appropriate management of concomitant drugs are highly recommended.

5.
Ann Noninvasive Electrocardiol ; 27(1): e12888, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34547155

RESUMEN

BACKGROUND: TdP is a form of polymorphic ventricular tachycardia which develops in the setting of a prolonged QT interval. There are limited data describing risk factors, treatment, and outcomes of this potentially fatal arrhythmia. OBJECTIVE: Our goals were as follows: (1) to validate cases presenting with Torsade de Pointes (TdP), (2) to identify modifiable risk factors, and (3) to describe the management strategies used for TdP and its prognosis in a real-world healthcare setting. METHODS: Case-control study (with 2:1 matching on age, sex, and race/ethnicity) nested within the Genetic Epidemiology Research on Aging (GERA) cohort. Follow-up of the cohort for case ascertainment was between January 01, 2005 and December 31, 2018. RESULTS: A total of 56 cases of TdP were confirmed (incidence rate = 3.6 per 100,000 persons/years). The average (SD) age of the TdP cases was 74 (13) years, 55 percent were female, and 16 percent were non-white. The independent predictors of TdP were potassium concentration <3.6 mEq/L (OR = 10.6), prior history of atrial fibrillation/flutter (OR = 6.2), QTc >480 ms (OR = 4.4) and prior history of coronary artery disease (OR = 2.6). Exposure to furosemide and amiodarone was significantly greater in cases than in controls. The most common treatment for TdP was IV magnesium (78.6%) and IV potassium repletion (73.2%). The in-hospital and 1-year mortality rates for TdP cases were 10.7% and 25.0% percent, respectively. CONCLUSIONS: These findings may inform quantitative multivariate risk indices for the prediction of TdP and could guide practitioners on which patients may qualify for continuous ECG monitoring and/or electrolyte replacement therapy.


Asunto(s)
Prestación Integrada de Atención de Salud , Síndrome de QT Prolongado , Torsades de Pointes , Anciano , Estudios de Casos y Controles , Electrocardiografía , Femenino , Humanos , Torsades de Pointes/diagnóstico , Torsades de Pointes/epidemiología
6.
Eur J Pharmacol ; 915: 174670, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34863995

RESUMEN

Hydroxychloroquine (HCQ) is a derivative of the antimalaria drug chloroquine primarily prescribed for autoimmune diseases. Recent attempts to repurpose HCQ in the treatment of corona virus disease 2019 has raised concerns because of its propensity to prolong the QT-segment on the electrocardiogram, an effect associated with increased pro-arrhythmic risk. Since chirality can affect drug pharmacological properties, we have evaluated the functional effects of the R(-) and S(+) enantiomers of HCQ on six ion channels contributing to the cardiac action potential and on electrophysiological parameters of isolated Purkinje fibers. We found that R(-)HCQ and S(+)HCQ block human Kir2.1 and hERG potassium channels in the 1 µM-100 µM range with a 2-4 fold enantiomeric separation. NaV1.5 sodium currents and CaV1.2 calcium currents, as well as KV4.3 and KV7.1 potassium currents remained unaffected at up to 90 µM. In rabbit Purkinje fibers, R(-)HCQ prominently depolarized the membrane resting potential, inducing autogenic activity at 10 µM and 30 µM, while S(+)HCQ primarily increased the action potential duration, inducing occasional early afterdepolarization at these concentrations. These data suggest that both enantiomers of HCQ can alter cardiac tissue electrophysiology at concentrations above their plasmatic levels at therapeutic doses, and that chirality does not substantially influence their arrhythmogenic potential in vitro.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Corazón/efectos de los fármacos , Hidroxicloroquina/química , Hidroxicloroquina/farmacología , Canales Iónicos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/inducido químicamente , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Canales de Potasio Éter-A-Go-Go , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Ramos Subendocárdicos/efectos de los fármacos , Conejos , Estereoisomerismo
7.
Circ Arrhythm Electrophysiol ; 14(3): e009458, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33554620
8.
Toxicol Appl Pharmacol ; 390: 114883, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31981640

RESUMEN

Human-based in silico models are emerging as important tools to study the effects of integrating inward and outward ion channel currents to predict clinical proarrhythmic risk. The aims of this study were 2-fold: 1) Evaluate the capacity of an in silico model to predict QTc interval prolongation in the in vivo anesthetized cardiovascular guinea pig (CVGP) assay for new chemical entities (NCEs) and; 2) Determine if a translational pharmacokinetic/pharmacodynamic (tPKPD) model can improve the predictive capacity. In silico simulations for NCEs were performed using a population of human ventricular action potential (AP) models. PatchXpress® (PX) or high throughput screening (HTS) ion channel data from respectively n = 73 and n = 51 NCEs were used as inputs for the in silico population. These NCEs were also tested in the CVGP (n = 73). An M5 pruned decision tree-based regression tPKPD model was used to evaluate the concentration at which an NCE is liable to prolong the QTc interval in the CVGP. In silico results successfully predicted the QTc interval prolongation outcome observed in the CVGP with an accuracy/specificity of 85%/73% and 75%/77%, when using PX and HTS ion channel data, respectively. Considering the tPKPD predicted concentration resulting in QTc prolongation (EC5%) increased accuracy/specificity to 97%/95% using PX and 88%/97% when using HTS. Our results support that human-based in silico simulations in combination with tPKPD modeling can provide correlative results with a commonly used early in vivo safety assay, suggesting a path toward more rapid NCE assessment with reduced resources, cycle time, and animal use.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas , Simulación por Computador , Técnicas Electrofisiológicas Cardíacas , Modelos Biológicos , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Línea Celular , Fenómenos Electrofisiológicos/efectos de los fármacos , Cobayas , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Modelos Químicos
9.
J Cardiol Cases ; 20(4): 132-134, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31969942

RESUMEN

Torsade de Pointes (TdP) is a rare cardiac arrhythmia that is associated with prolonged QTc interval. Hypocalcemia is a common cause of prolonged QTc. Although vitamin D deficiency (VDD) is a common disorder in elderly patients with an incidence rate of >40% and can cause hypocalcemia, it has never been linked to TdP. We report a patient with severe VDD that resulted in TdP and cardiac arrest. Post-resuscitation work up illustrated prolonged QTc interval of 620 ms, significant hypocalcemia, and severe VDD of 4 (normal 30-80) ng/mL. After high dose vitamin D/calcium supplements, repeat electrocardiogram revealed normal QTc interval of 423 ms. During hospitalization, the patient suffered no additional arrhythmias and QTc continued to be normal. .

10.
Ther Innov Regul Sci ; 53(4): 519-525, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30157676

RESUMEN

A Cardiac Safety Research Consortium / Health and Environmental Sciences Institute / FDA-sponsored Think Tank Meeting was convened in Washington, DC, on May 21, 2018, to bring together scientists, clinicians, and regulators from multiple geographic regions to evaluate progress to date in the Comprehensive In Vitro Proarrhythmia Assay (CiPA) Initiative, a new paradigm to evaluate proarrhythmic risk. Study reports from the 4 different components of the CiPA paradigm (ionic current studies, in silico modeling to generate a Torsade Metric Score, human induced pluripotent stem cell-derived ventricular cardiomyocytes, and clinical ECG assessments including J-Tpeakc) were presented and discussed. This paper provides a high-level summary of the CiPA data presented at the meeting.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Bioensayo , Simulación por Computador , Electrocardiografía , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Canales Iónicos/fisiología , Miocitos Cardíacos
11.
Cell Rep ; 24(13): 3582-3592, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30257217

RESUMEN

To assess the utility of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as an in vitro proarrhythmia model, we evaluated the concentration dependence and sources of variability of electrophysiologic responses to 28 drugs linked to low, intermediate, and high torsades de pointes (TdP) risk categories using two commercial cell lines and standardized protocols in a blinded multisite study using multielectrode array or voltage-sensing optical approaches. Logistical and ordinal linear regression models were constructed using drug responses as predictors and TdP risk categories as outcomes. Three of seven predictors (drug-induced arrhythmia-like events and prolongation of repolarization at either maximum tested or maximal clinical exposures) categorized drugs with reasonable accuracy (area under the curve values of receiver operator curves ∼0.8). hiPSC-CM line, test site, and platform had minimal influence on drug categorization. These results demonstrate the utility of hiPSC-CMs to detect drug-induced proarrhythmic effects as part of the evolving Comprehensive In Vitro Proarrhythmia Assay paradigm.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Electrofisiología/métodos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/efectos de los fármacos , Torsades de Pointes/inducido químicamente , Cardiotoxicidad , Línea Celular , Reprogramación Celular , Evaluación Preclínica de Medicamentos/normas , Electrofisiología/normas , Humanos , Potenciales de la Membrana/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología
12.
Artículo en Inglés | MEDLINE | ID: mdl-29958940

RESUMEN

The Safety Pharmacology Society organized a scientific session at its annual conference in 2017 to discuss the challenges and opportunities of the Comprehensive In-Vitro Proarrhythmia Assay (CiPA) paradigm. Our intention was to raise awareness of this initiative with its members and also to gauge the extent to which safety pharmacologists have incorporated the CiPA testing strategy within the pharmaceutical industry. CiPA offers many potential opportunities including 1) a focus on proarrhythmic risk (as opposed to QTc prolongation), 2) providing scientific rationale to support the continued development of compounds that may have a poor selectivity over hERG whilst also blocking other inward currents and 3) reducing the extent of ECG monitoring in clinical trials with a greater influence of the non-clinical studies. Such opportunities may speed drug development and reduce costs. However, there are also challenges for CiPA implementation. For example, the mixed ion channel paradigm does not easily lend itself to a prospective drug discovery strategy although testing for such effects can be achieved with assays with good throughput. However, it should also be recognized that compounds with a mixed ion channel profile might also have properties that are undesirable to treat non-life threatening indications. All components of CiPA (nonclinical and clinical) require validation, particularly as a composite package to impact drug development and evaluation. One of the significant discussion points was that the existing regulatory guidance supports the use of components of CiPA through follow-up studies. A survey of the conference audience showed that the level of awareness of CiPA is quite high and that companies are already conducting some testing against a wider panel of cardiac ion channels beyond hERG. However, the adoption of other technologies (stem cell derived cardiac myocytes and in silico modeling) is less well developed. Taken together, the session demonstrated the potential advantages of CiPA, but also some significant challenges.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Simulación por Computador , Congresos como Asunto , Electrocardiografía/efectos de los fármacos , Sociedades Farmacéuticas , Animales , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/prevención & control , Simulación por Computador/normas , Congresos como Asunto/normas , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Canal de Potasio ERG1/agonistas , Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/fisiología , Electrocardiografía/normas , Humanos , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/fisiopatología , Síndrome de QT Prolongado/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Sociedades Farmacéuticas/normas
13.
J Pharmacokinet Pharmacodyn ; 45(3): 457-467, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29520534

RESUMEN

Cardiotoxicity is among the top drug safety concerns, and is of specific interest in tuberculosis, where this is a known or potential adverse event of current and emerging treatment regimens. As there is a need for a tool, beyond the QT interval, to quantify cardiotoxicity early in drug development, an empirical decision tree based classifier was developed to predict the risk of Torsades de pointes (TdP). The cardiac risk algorithm was developed using pseudo-electrocardiogram (ECG) outputs derived from cardiac myocyte electromechanical model simulations of increasing concentrations of 96 reference compounds which represented a range of clinical TdP risk. The algorithm correctly classified 89% of reference compounds with moderate sensitivity and high specificity (71 and 96%, respectively) as well as 10 out of 12 external validation compounds and the anti-TB drugs moxifloxacin and bedaquiline. The cardiac risk algorithm is suitable to help inform early drug development decisions in TB and will evolve with the addition of emerging data.


Asunto(s)
Antituberculosos/efectos adversos , Antituberculosos/uso terapéutico , Cardiotoxicidad/etiología , Corazón/efectos de los fármacos , Torsades de Pointes/inducido químicamente , Tuberculosis/tratamiento farmacológico , Adulto , Algoritmos , Diarilquinolinas/efectos adversos , Diarilquinolinas/uso terapéutico , Desarrollo de Medicamentos/métodos , Electrocardiografía/métodos , Femenino , Humanos , Masculino , Moxifloxacino/efectos adversos , Medición de Riesgo , Sensibilidad y Especificidad
14.
J Electrocardiol ; 51(1): 111-116, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29037423

RESUMEN

A 26-year-old woman, 12 days in postpartum, developed recurrent syncope and cardiac arrest. Her ECG revealed QT-prolongation associated with LQT2-specific T-U wave patterns, T wave alternans, long QT-dependent torsade de pointes (TdP) and ventricular fibrillation (VF). She also had intermittent LBBB (80bpm) on alternate beats and RBBB at sinus tachycardia (113bpm). Family genotyping revealed a novel de novo missense mutation G604C of KCNH2. Propranolol slowed heart rate and further prolonged QT interval (610ms) that caused TdP recurrence. Mexiletine combined with magnesium and potassium supplements prevented TdP/VF recurrence. This patient has remained event-free after 9-month follow-up.


Asunto(s)
Electrocardiografía , Síndrome de QT Prolongado/genética , Mutación Missense , Antagonistas Adrenérgicos beta/efectos adversos , Adulto , Femenino , Genotipo , Frecuencia Cardíaca/efectos de los fármacos , Heterocigoto , Humanos , Síndrome de QT Prolongado/complicaciones , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/tratamiento farmacológico , Periodo Posparto , Síncope/etiología , Torsades de Pointes/etiología , Fibrilación Ventricular/etiología
15.
China Pharmacist ; (12): 328-330, 2018.
Artículo en Chino | WPRIM | ID: wpr-705523

RESUMEN

Objective:To analyze the general patterns and characteristics of torsade de pointes(Tdp) associated with ibutilide and provide feasible suggestions and preventive measurements combined with the experience of pharmaceutical monitoring. Methods: The cases of TdP associated with ibutilide were retrieved from the literatures reported at home and abroad during January 1990 and April 2017,and the clinical data including gender,age,original diseases, dosage of drugs, injection time, TdP occurrence time, potassi-um,QTc and ECG monitoring before and after the medication, complications, treatment drugs and methods and conversion outcome were statistically analyzed. Results: A total of 9 cases were enrolled in the analysis. After the treatment with ibutilide withdrawal, electroversion and potassium and magnesium supplement, all the patients converted to sinus rhythm.Conclusion: Ibutilide must be prescribed very carefully in senile female patients complicated with extended QTc,low potassium,organic heart disease and heart fail-ure. With dosing interval,injection time or injection speed not compatible with the specification,the occurrence of TdP is significantly different,suggesting that clinical pharmacists strengthen the monitoring of injection time,speed,dosing interval and blood concentra-tion of ibutilide according to the instruction strictly.

16.
J Pharmacol Toxicol Methods ; 87: 11-23, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28408211

RESUMEN

Voltage gated ion channels are central in defining the fundamental properties of the ventricular cardiac action potential (AP), and are also involved in the development of drug-induced arrhythmias. Many drugs can inhibit cardiac ion currents, including the Na+ current (INa), L-type Ca2+ current (Ica-L), and K+ currents (Ito, IK1, IKs, and IKr), and thereby affect AP properties in a manner that can trigger or sustain cardiac arrhythmias. Since publication of ICH E14 and S7B over a decade ago, there has been a focus on drug effects on QT prolongation clinically, and on the rapidly activating delayed rectifier current (IKr), nonclinically, for evaluation of proarrhythmic risk. This focus on QT interval prolongation and a single ionic current likely impacted negatively some drugs that lack proarrhythmic liability in humans. To rectify this issue, the Comprehensive in vitro proarrhythmia assay (CiPA) initiative has been proposed to integrate drug effects on multiple cardiac ionic currents with in silico modelling of human ventricular action potentials, and in vitro data obtained from human stem cell-derived ventricular cardiomyocytes to estimate proarrhythmic risk of new drugs with improved accuracy. In this review, we present the physiological functions and the molecular basis of major cardiac ion channels that contribute to the ventricle AP, and discuss the CiPA paradigm in drug development.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Cardiotoxinas/farmacología , Canales Iónicos/fisiología , Farmacología/métodos , Animales , Cardiotoxinas/efectos adversos , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiología , Humanos , Canales Iónicos/agonistas , Canales Iónicos/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Farmacología/normas
17.
Artículo en Inglés | MEDLINE | ID: mdl-28163191

RESUMEN

INTRODUCTION: The use of multi-electrode arrays (MEA) in combination with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provides a promising method to predict comprehensive cardiotoxicity, including drug-induced QT prolongation and arrhythmia. We previously demonstrated that MEA in combination with hiPSC-CMs could provide a generalizable platform by using 7 reference drugs at 10 testing facilities. Using this approach, we evaluated responses to reference drugs that modulate a range of cardiac ion currents and have a range of arrhythmogenic effects. METHODS: We used the MEA system (MED64) and commercially available hiPSC-CMs (iCell cardiomyocytes) to evaluate drug effects on the beat rate, field potential duration (FPD), FPD corrected by Fridericia's formula (FPDc), and the incidence of arrhythmia-like waveforms. RESULTS: This assay detected the repolarization effects of Bay K8644, mibefradil, NS1643, levcromakalim, and ouabain; and the chronotropic effects of isoproterenol, ZD7288, and BaCl2. Chronotropy was also affected by K+ and Ca2+ current modulation. This system detected repolarization delays and the arrhythmogenic effects of quinidine, cisapride, thioridazine, astemizole, bepridil, and pimozide more sensitively than the established guinea pig papillary muscle action potential assay. It also predicted clinical QT prolongation by drugs with multiple ion channel effects (fluoxetine, amiodarone, tolterodine, vanoxerine, alfuzosin, and ranolazine). DISCUSSION: MEA in combination with hiPSC-CMs may provide a powerful method to detect various cardiac electrophysiological effects, QT prolongation, and arrhythmia during drug discovery. However, the data require careful interpretation to predict chronotropic effects and arrhythmogenic effects of candidate drugs with multiple ion channel effects.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Cardiotoxinas/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Canales Iónicos , Miocitos Cardíacos/efectos de los fármacos , Arritmias Cardíacas/fisiopatología , Cardiotónicos/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Frecuencia Cardíaca/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Canales Iónicos/agonistas , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/fisiología , Miocitos Cardíacos/fisiología
18.
Heart Rhythm ; 14(5): 749-756, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28213055

RESUMEN

BACKGROUND: Pacing at higher rates is known to suppress torsade de pointes (TdP) arrhythmias. Nevertheless, exact application and mechanism need further clarification. In the anesthetized canine chronic atrioventricular block model, ventricular remodeling is responsible for a high and reproducible incidence of TdP upon a challenge with dofetilide. OBJECTIVE: We used this model to investigate by what mechanism accelerated pacing averts TdP and what repolarization parameter could be used to guide temporary accelerated pacing (TAP). METHODS: Ten dogs with repetitive TdP after administration of dofetilide when paced at 60 beats/min were selected. In a serial experiment, TAP was initiated at 100 beats/min after the first ectopic beat. Electrocardiogram and right and left ventricular (LV) monophasic action potential durations (MAPDs) were recorded. In a subset, vertical dispersion was determined with a duodecapolar catheter. Temporal dispersion was quantified as short-term variability (STV). Arrhythmias were quantified with the arrhythmia score. RESULTS: The increase in repolarization parameters observed after administration of dofetilide was counteracted by TAP (eg, LV MAPD from 381 ± 94 ms back to 310 ± 17 ms; P < .05). Temporal dispersion (STVLVMAPD) increased from 0.69 ± 0.37 to 2.59 ± 0.96 ms (P < .05) after administration of dofetilide and back to 1.15 ± 0.54 ms (P < .05) with TAP. This was accompanied by suppression of recurrent TdP in 7 of 10 dogs (P < .05) and a trend toward reduction in vertical (spatial) dispersion from 56 ± 25 to 31 ± 4 ms (P = .06). In those dogs, seconds after capture of TAP, almost all ectopy disappeared, causing a decrease in arrhythmia score from 21 ± 12 to 4 ± 3 (P < .05). CONCLUSION: TAP is effective in averting TdP by decreasing spatial and temporal measures of repolarization. Increase in temporal dispersion (STV) can guide TAP.


Asunto(s)
Bloqueo Atrioventricular/fisiopatología , Bloqueo Atrioventricular/terapia , Estimulación Cardíaca Artificial/métodos , Torsades de Pointes/prevención & control , Torsades de Pointes/fisiopatología , Animales , Bloqueo Atrioventricular/complicaciones , Enfermedad Crónica , Modelos Animales de Enfermedad , Perros , Técnicas Electrofisiológicas Cardíacas , Fenómenos Electrofisiológicos , Frecuencia Cardíaca/fisiología , Recurrencia , Torsades de Pointes/etiología , Remodelación Ventricular/fisiología
19.
JACC Clin Electrophysiol ; 3(13): 1565-1576, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29759839

RESUMEN

OBJECTIVES: This study investigated the arrhythmogenic mechanisms responsible for torsade de pointes (TdP) in the chronic atrioventricular block dog model, known for its high susceptibility for TdP. BACKGROUND: The mechanism of TdP arrhythmias has been under debate for many years. Focal activity as well as re-entry have both been mentioned in the initiation and the perpetuation of TdP. METHODS: In 5 TdP-sensitive chronic atrioventricular block dogs, 56 needle electrodes were evenly distributed transmurally to record 240 unipolar local electrograms simultaneously. Nonterminating (NT) episodes were defibrillated after 10 s. Software was developed to automatically detect activation times and to create 3-dimensional visualizations of the arrhythmia. For each episode of ectopic activity (ranging from 2 beats to NT episodes), a novel methodology was created to construct directed graphs of the wave propagation and detect re-entry loops by using an iterative depth-first-search algorithm. RESULTS: Depending on the TdP definition (number of consecutive ectopic beats), we analyzed 29 to 54 TdP: 29 were longer than 5 beats. In the total group, 9 were NT and 45 were self-terminating. Initiation and termination were always based on focal activity. Re-entry becomes more important in the longer-lasting episodes (>14 beats), whereas in all NT TdP, re-entry was the last active mechanism. During re-entry, excitation fronts were constantly present in the heart, while during focal TdP, there was always a silent interval between 2 consecutive waves (142 ms) during which excitation fronts were absent. Interbeat intervals were significantly smaller for re-entry episodes-220 versus 310 ms in focal. Electrograms recorded in particular areas during NT TdP episodes had significantly smaller amplitude (0.38) than during focal episodes (0.59). CONCLUSIONS: TdP can be driven by focal activity as well as by re-entry depending on the duration of the episode. NT episodes are always maintained by re-entry, which can be identified in local unipolar electrograms by shorter interbeat intervals and smaller deflection amplitude.


Asunto(s)
Bloqueo Atrioventricular/fisiopatología , Electrodos Implantados/estadística & datos numéricos , Técnicas Electrofisiológicas Cardíacas/instrumentación , Torsades de Pointes/fisiopatología , Algoritmos , Animales , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/fisiopatología , Bloqueo Atrioventricular/veterinaria , Perros , Electrocardiografía/métodos , Electrodos Implantados/efectos adversos , Modelos Animales , Países Bajos/epidemiología
20.
Int J Cardiol ; 214: 218-24, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27064645

RESUMEN

INTRODUCTION: Ischemia causes myocardial infarction and arrhythmias. Up-regulation of cardiac CLC-3 chloride channels is important for ischemic preconditioning-induced second-window protection against myocardial infarction. But its consequences in ischemia-induced electrical remodeling are still unknown. METHODS: The recently-characterized heart-specific overexpression of human short CLC-3 isoform (hsCLC-3(OE)) mice was used to study the effects of CLC-3 up-regulation on cardiac electrophysiology under ischemia/reperfusion conditions. In vivo surface electrocardiography (ECG) and intracardiac electrophysiology (ICEP) were used to compare the electrophysiological properties of age-matched wild-type (Clcn3(+/+)) and hsCLC-3(OE) mice under control and myocardial ischemia-reperfusion conditions. RESULTS: QT and QTc intervals of hsCLC-3(OE) mice were significantly shorter than those of Clcn3(+/+) mice under control, ischemia and reperfusion conditions. In the ICEP, ventricular effective refractory period (VERP) of hsCLC-3(OE) mice (26.7±1.7ms, n=6) was significantly shorter than that of Clcn3(+/+) mice (36.9±2.8ms, n=8, P<0.05). Under ischemia condition, both VERP (19.8±1.3ms) and atrial effective refractory period (AERP, 34.8±2.5ms) of hsCLC-3(OE) mice were significantly shorter than those of Clcn3(+/+) mice (35.2±3.0ms and 45.8±1.6ms, P<0.01, respectively). Wenckebach atrioventricular nodal block point (AVBP, 91.13±4.08ms) and 2:1 AVBP (71.3±3.8ms) of hsCLC-3(OE) mice were significantly shorter than those of Clcn3(+/+) mice (102.0±2.0ms and 84.1±2.8ms, P<0.05, respectively). However, no differences of ICEP parameters between hsCLC-3(OE) and Clcn3(+/+) mice were observed under reperfusion conditions. CONCLUSION: Heart-specific overexpression of hsCLC-3 limited the ischemia-induced QT and ERP prolongation and postponed the advancements of Wenckebach and 2:1 AVBP. CLC-3 up-regulation may serve as an important adaptive mechanism against myocardial ischemia.


Asunto(s)
Canales de Cloruro/metabolismo , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Modelos Animales de Enfermedad , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/fisiopatología , Isoformas de Proteínas/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA