Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 800
Filter
1.
Clin Pharmacol Drug Dev ; 13(6): 590-600, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38623935

ABSTRACT

This open-label, phase 1 study was conducted with healthy adult participants to evaluate the potential drug-drug interaction between rilzabrutinib and quinidine (an inhibitor of P-glycoprotein [P-gp] and CYP2D6) or rifampin (an inducer of CYP3A and P-gp). Plasma concentrations of rilzabrutinib were measured after a single oral dose of rilzabrutinib 400 mg administered on day 1 and again, following a wash-out period, after co-administration of rilzabrutinib and quinidine or rifampin. Specifically, quinidine was given at a dose of 300 mg every 8 hours for 5 days from day 7 to day 11 (N = 16) while rifampin was given as 600 mg once daily for 11 days from day 7 to day 17 (N = 16) with rilzabrutinib given in the morning of day 10 (during quinidine dosing) or day 16 (during rifampin dosing). Quinidine had no significant effect on rilzabrutinib pharmacokinetics. Rifampin decreased rilzabrutinib exposure (the geometric mean of Cmax and AUC0-∞ decreased by 80.5% and 79.5%, respectively). Single oral doses of rilzabrutinib, with or without quinidine or rifampin, appeared to be well tolerated. These findings indicate that rilzabrutinib is a substrate for CYP3A but not a substrate for P-gp.


Subject(s)
Area Under Curve , Drug Interactions , Healthy Volunteers , Quinidine , Rifampin , Humans , Rifampin/administration & dosage , Rifampin/adverse effects , Quinidine/administration & dosage , Quinidine/adverse effects , Quinidine/pharmacology , Quinidine/pharmacokinetics , Adult , Male , Female , Young Adult , Middle Aged , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inducers/adverse effects , Cytochrome P-450 CYP3A/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Pyrimidines/adverse effects
2.
Clin Pharmacol Ther ; 115(3): 616-626, 2024 03.
Article in English | MEDLINE | ID: mdl-38117225

ABSTRACT

The lethality of torsades de pointes (TdP) by drugs is one of main reasons that some drugs were withdrawn from the market. In order to assess drug-induced TdP risks, a model of cardiac ionic current suppression in human ventricular myocytes (ToR-ORd model), combined with the maximum effective free therapeutic plasma concentration or the maximum effective free therapeutic myocyte concentration was often used, with the latter proved to be more relevant and more accurate. We aimed to develop a whole-body physiologically-based pharmacokinetic (PBPK) model, incorporated with a human cardiomyocyte pharmacodynamic (PD) model, to provide a comprehensive assessment of drug-induced TdP risks in normal and specific scenarios. Quinidine served as an example to validate the PBPK-PD model via predicting plasma quinidine concentrations and quinidine-induced changes in QT interval (ΔQTc). The predicted plasma quinidine concentrations and ΔQTc values following oral administration or intravenous administration of quinidine were comparable to clinic observations. Visual predictive checks showed that most of the observed plasma concentrations and ΔQTc values fell within the 5th and 95th percentiles of simulations. The validated PBPK-PD model was further applied to assess the TdP risks using frequencies of early afterdepolarization and long-QT syndrome occurrence in 4 scenarios, such as therapeutic dose, supra-therapeutic dose, alkalosis, and hyperkalemia in 200 human subjects. In conclusion, the developed PBPK-PD model may be applied to predict the quinidine pharmacokinetics and quinidine-induced TdP risks in healthy subjects, but also simulate quinidine-induced TdP risks under disease conditions, such as hypokalemia and alkalosis.


Subject(s)
Alkalosis , Long QT Syndrome , Torsades de Pointes , Humans , Quinidine/adverse effects , Torsades de Pointes/drug therapy , Electrocardiography , Long QT Syndrome/drug therapy , Alkalosis/drug therapy , DNA-Binding Proteins/therapeutic use
3.
J Clin Psychopharmacol ; 43(5): 422-427, 2023.
Article in English | MEDLINE | ID: mdl-37683231

ABSTRACT

BACKGROUND: Approximately one third of individuals with major depressive disorder have treatment-resistant depression (TRD). Glutamatergic modulators such as the N -methyl- d -aspartate receptor antagonist ketamine have rapid and robust antidepressant effects, but their use has been limited by accessibility and route of administration. This open-label pilot study assessed the adjunctive antidepressant efficacy of dextromethorphan/quinidine (DM/Q) in TRD. METHODS: Inpatients with TRD (n = 17, 40.8 ± 12.3 years; 9 females/8 males) received adjunctive open-label DM/Q (20 mg/10 mg) up to 3 times daily. The study had no set endpoint; participants were followed until they discontinued DM/Q or were discharged. Montgomery-Asberg Depression Rating Scale (MADRS) scores were obtained at baseline (before DM/Q administration) and regularly during hospitalization. Full response was defined as a ≥50% reduction in baseline MADRS score, partial response as a 25% to 50% decrease in baseline MADRS score, and nonresponse as a <25% reduction or an increase in baseline MADRS score. RESULTS: The 17 inpatients received open-label DM/Q for 5.1 ± 2.7 weeks. Forty-seven percent of participants responded to DM/Q-12% achieved a full response and 35% achieved a partial response. The largest MADRS difference observed at any time point was -6.4 ± 8.4 (-21.0% ± 29.9%), and the MADRS difference observed at time of DM/Q discontinuation or hospital discharge was -4.8 ± 8.4 (-15.9% ± 29.7%). Twenty-four percent of participants experienced a nonserious adverse event; none experienced a serious adverse event. CONCLUSIONS: In this open-label pilot study, 47% of participants responded to adjunctive DM/Q, which was well tolerated. Larger placebo-controlled trials are needed to determine the real-world efficacy of DM/Q.


Subject(s)
Depressive Disorder, Major , Depressive Disorder, Treatment-Resistant , Male , Female , Humans , Quinidine/adverse effects , Dextromethorphan/pharmacology , Depressive Disorder, Major/drug therapy , Treatment Outcome , Depression , Pilot Projects , Antidepressive Agents/adverse effects , Depressive Disorder, Treatment-Resistant/drug therapy , Double-Blind Method
4.
Br J Clin Pharmacol ; 88(12): 5096-5112, 2022 12.
Article in English | MEDLINE | ID: mdl-35940594

ABSTRACT

AIMS: Despite numerous studies on quinidine therapies for epilepsies associated with KCNT1 gene mutations, there is no consensus on its clinical utility. Thus, we reviewed studies evaluating the efficacy and safety of quinidine in KCNT1-related epileptic disorders. METHODS: Electronic databases were queried for in vivo and in vitro studies on quinidine therapy in KCNT1-related epilepsies published on or before 1 May 2022. The evaluation of evidence was done as per the American Academy of Neurology's classification scheme. Identification of significant factors that possibly influenced therapeutic effects of quinidine were performed using χ2 tests. RESULTS: Twenty-seven studies containing 82 patient records were reviewed. Records of 80 patients with 33 KCNT1 mutations were analysed, of which 20 patients had gained ≥50% seizure reduction due to quinidine therapy. However, quinidine therapy often had different effects on patients with the same KCNT1 mutation. Age, genotypes of KCNT1 mutations, seizure types and brain MRI did not significantly influence the therapeutic effect of quinidine. Prolonged QTc was the most common among all adverse events with quinidine. Notably, results of in vitro quinidine tests did not correspond with in vivo tests. CONCLUSIONS: Therapeutic effects of quinidine on KCNT1-related epilepsies remained indefinite as contradictory results were detected in similar patients. Age, seizure types, genotypes of KCNT1 mutations and brain MRI did not influence the therapeutic effects of quinidine. Insensitivity to quinidine by a certain Kcnt1 genotype in molecular tests is predictive of its inefficacy in human populations of the respective mutation.


Subject(s)
Epilepsy , Quinidine , Humans , Quinidine/adverse effects , Potassium Channels, Sodium-Activated/genetics , Anticonvulsants/adverse effects , Nerve Tissue Proteins , Epilepsy/drug therapy , Epilepsy/genetics , Seizures/drug therapy , Mutation
5.
Exp Physiol ; 107(8): 864-878, 2022 08.
Article in English | MEDLINE | ID: mdl-35561081

ABSTRACT

NEW FINDINGS: What is the central question of this study? Can the triangular appearance of ventricular action potential, indicating proarrhythmic profile of antiarrhythmic agent, be approximated by specific changes on an electrocardiogram (ECG)? What are the main finding and its importance? The triangulation of the ventricular action potential seen when antiarrhythmic drugs induce a greater lengthening of the late repolarization compared to the initial repolarization in epicardium is closely approximated by a greater prolongation of the T wave upslope relative to the interval between the J point and the start of the T wave (the JTstart interval) on the ECG. These findings may improve the power of ECG assessments in predicting the drug-induced arrhythmia resulting from slowed phase 3 repolarization. ABSTRACT: Antiarrhythmic drugs prescribed to treat atrial fibrillation can occasionally precipitate ventricular tachyarrhythmia through a prominent slowing of the phase 3 repolarization. The latter results in the triangular shape of ventricular action potential, indicating high arrhythmic risk. However, clinically, the utility of triangulation assessments for predicting arrhythmia is limited owing to the invasive nature of the ventricular action potential recordings. This study examined whether the triangulation effect can be detected indirectly from electrocardiogram (ECG) analysis. Epicardial monophasic action potentials and the ECG were simultaneously recorded in perfused guinea-pig hearts. With antiarrhythmics (dofetilide, quinidine, procainamide and flecainide), a prolongation of the initial repolarization seen in the action potential recordings was closely approximated by lengthening of the interval between the J point and the start of the T wave (the JTstart interval) on the ECG, whereas a prolongation of the late repolarization was paralleled by widening of the T wave upslope. Dofetilide, quinidine and procainamide induced a prominent slowing of the phase 3 repolarization in epicardium, leading to triangulation of the action potential. These effects were accompanied by a greater prolongation of the T wave upslope compared to the JTstart interval. Flecainide elicited a proportional prolongation of the initial and the late ventricular repolarization, and therefore failed to induce triangulation, based on analysis of both epicardial action potential and ECG profiles. Collectively, these findings suggest that the ratio between the durations of the T wave upslope and the JTstart interval may represent the ECG metric of the ventricular action potential triangulation induced by antiarrhythmic drugs.


Subject(s)
Anti-Arrhythmia Agents , Arrhythmias, Cardiac , Action Potentials , Animals , Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/drug therapy , Electrocardiography , Flecainide/adverse effects , Guinea Pigs , Pericardium/drug effects , Pericardium/physiology , Procainamide/adverse effects , Quinidine/adverse effects
6.
J Cardiovasc Electrophysiol ; 32(12): 3173-3178, 2021 12.
Article in English | MEDLINE | ID: mdl-34586686

ABSTRACT

INTRODUCTION: Quinidine is an effective therapy for a subset of polymorphic ventricular tachycardia and ventricular fibrillation (VF) syndromes; however, the efficacy of quinidine in scar-related monomorphic ventricular tachycardia (MMVT) is unclear. METHODS AND RESULTS: Between 2009 and 2020 a single VT referral center, a total of 23 patients with MMVT and structural heart disease (age 66.7 ± 10.9, 20 males, 15 with ischemic cardiomyopathy, mean LVEF 22.2 ± 12.3%, 9 with left ventricular assist device [LVAD]) were treated with quinidine (14 quinidine gluconate; 996 ± 321 mg, 8 quinidine sulfate; 1062 ± 588 mg). Quinidine was used in combination with other antiarrhythmics (AAD) in 19 (13 also on amiodarone). All patients previously failed >1 AAD (amiodarone 100%, mexiletine 73%, sotalol 32%, other 32%) and eight had prior ablations (median of 1.5). Quinidine was initiated in the setting of VT storm despite AADs (6), inability to tolerate other AADs (4), or recurrent VT(12). Ventricular arrhythmias recurred despite quinidine in 13 (59%) patients at a median of 26 (4-240) days after quinidine initiation. In patients with recurrent MMVT, VT cycle length increased from 359 to 434 ms (p = .02). Six (27.3%) patients remained on quinidine at 1 year with recurrence of ventricular arrhythmias in all. The following adverse effects were seen: gastrointestinal side effects (6), QT prolongation (2), rash (1), thrombocytopenia (1), neurologic side effects (1). One patient discontinued due to cost. CONCLUSION: Quinidine therapy has limited tolerability and long-term efficacy when used in the management of amiodarone-refractory scar-related MMVT.


Subject(s)
Quinidine , Tachycardia, Ventricular , Anti-Arrhythmia Agents/adverse effects , Humans , Male , Quinidine/adverse effects , Salvage Therapy , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/drug therapy , Ventricular Fibrillation
7.
SLAS Discov ; 26(3): 364-372, 2021 03.
Article in English | MEDLINE | ID: mdl-32914673

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been widely used for the assessment of drug proarrhythmic potential through multielectrode array (MEA). HiPSC-CM cultures beat spontaneously with a wide range of frequencies, however, which could affect drug-induced changes in repolarization. Pacing hiPSC-CMs at a physiological heart rate more closely resembles the state of in vivo ventricular myocytes and permits the standardization of test conditions to improve consistency. In this study, we systematically investigated the time window of stable ion currents in high-purity hiPSC-derived ventricular cardiomyocytes (hiPSC-vCMs) and confirmed that these cells could be used to correctly predict the proarrhythmic risk of Comprehensive In Vitro Proarrhythmia Assay (CiPA) reference compounds. To evaluate drug proarrhythmic potentials at a physiological beating rate, we used a MEA to electrically pace hiPSC-vCMs, and we recorded regular field potential waveforms in hiPSC-vCMs treated with DMSO and 10 CiPA reference drugs. Prolongation of field potential duration was detected in cells after exposure to high- and intermediate-risk drugs; in addition, drug-induced arrhythmia-like events were observed. The results of this study provide a simple and feasible method to investigate drug proarrhythmic potentials in hiPSC-CMs at a physiological beating rate.


Subject(s)
Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/drug effects , Phenethylamines/adverse effects , Quinidine/adverse effects , Sulfonamides/adverse effects , Action Potentials/physiology , Arrhythmias, Cardiac/prevention & control , Calcium/metabolism , Cations, Divalent , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Ion Transport/drug effects , Microelectrodes , Models, Biological , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Nifedipine/pharmacology , Patch-Clamp Techniques , Primary Cell Culture , Sotalol/adverse effects , Tetrodotoxin/antagonists & inhibitors , Tetrodotoxin/toxicity , Verapamil/pharmacology
8.
BMJ Case Rep ; 13(8)2020 Aug 24.
Article in English | MEDLINE | ID: mdl-32843415

ABSTRACT

Quinidine is one of the oldest antiarrhythmics known. Over the years, its use has decreased along with its side effects. Our case describes a 69-year-old woman with recurrent resistant ventricular tachycardia on Quinidine and Amiodarone who presented with acute liver toxicity. Drug-induced liver toxicity was at the top of our differential diagnosis list. Taking multiple factors into consideration, a decision was made to discontinue Quinidine, the patient's symptoms and lab abnormalities resolved within 1 week, yielding the diagnosis of Quinidine hypersensitivity.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Chemical and Drug Induced Liver Injury , Drug Hypersensitivity/diagnosis , Quinidine/adverse effects , Acute Disease , Aged , Amiodarone/therapeutic use , Anti-Arrhythmia Agents/therapeutic use , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/etiology , Female , Humans , Quinidine/therapeutic use , Tachycardia, Ventricular/drug therapy
9.
Neuropsychol Rev ; 30(1): 28-50, 2020 03.
Article in English | MEDLINE | ID: mdl-31942705

ABSTRACT

Pseudobulbar affect is a debilitating condition that significantly reduces quality of life for many individuals following traumatic brain injury (TBI). It is characterized by embarrassing and often uncontrollable episodes of crying or laughter. The aim of this systematic review was to evaluate the effectiveness of pharmacotherapy as compared to all other comparators for the management of pseudobulbar affect in adults who have sustained TBI. Six databases were searched, with additional hand searching of journals, clinical trials registries and international drug regulators to identify published and unpublished studies in English up to June 2018. Studies were eligible for this review if they included adults who had sustained a medically confirmed TBI and presented with pseudobulbar affect. All pharmacotherapy and comparator interventions were considered for inclusion, and study design was not limited to randomised controlled trials. Evidence quality was assessed using Joanna Briggs Institute Critical Appraisal Instruments. Two quasi-experimental studies examining the effectiveness of dextrometamorphan/quinidine (DM/Q) were identified. These studies reported that DM/Q was effective in reducing symptoms of pseudobulbar affect and had a positive safety profile, over follow-up periods of 3 months (n = 87) and 12 months (n = 23). However, both studies were limited by lack of a control group and a high dropout rate. The findings of twelve case reports examining the effectiveness of DM/Q (n = 6) and anti-depressants (n = 6) are also discussed. Further research is required to determine which pharmacological interventions provide the best outcomes for individuals with pseudobulbar affect following TBI, with consideration given to side effect profiles and financial costs.


Subject(s)
Affective Symptoms/drug therapy , Affective Symptoms/etiology , Brain Injuries, Traumatic/complications , Dextromethorphan/pharmacology , Neurotransmitter Agents/pharmacology , Quinidine/pharmacology , Dextromethorphan/adverse effects , Drug Combinations , Humans , Neurotransmitter Agents/adverse effects , Quinidine/adverse effects
10.
Epileptic Disord ; 21(1): 48-54, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30782581

ABSTRACT

Several recent studies have reported potassium sodium-activated channel subfamily T member 1 (KCNT1) mutations in epilepsy patients on quinidine therapy. The efficacy and safety of quinidine for epilepsy treatment, however, remains controversial. We herein report the cases of four patients with KCNT1 mutations treated with quinidine. A reduction in seizures of more than 50% after quinidine treatment was observed in one patient with epilepsy of infancy with migrating focal seizures (EIMFS), whereas two patients with EIMFS and one with focal epilepsy did not achieve apparent seizure reduction. The relationship between quinidine dose and serum quinidine concentration was inconsistent, particularly at high quinidine doses. One patient with EIMFS developed ventricular tachycardia the day after an increase in quinidine dose from 114 to 126 mg/kg/day. The serum trough quinidine concentration and the corrected QT interval (QTc) before arrhythmia onset were 2.4 µg/ml and 420 ms, respectively, and peak serum quinidine concentration after arrhythmia onset was 9.4 µg/ml. Another patient with EIMFS showed aberrant intraventricular conduction with a quinidine dose of 74.5 mg/kg/day and a serum trough concentration of 3.2 µg/ml. Given that serum quinidine levels may elevate sharply after a dose increase, careful monitoring of electrocardiographs and serum concentrations is required. Based on a review of previous reports and our experience with this case, quinidine should be considered as a promising drug for patients with EIMFS harbouring KCNT1 mutations, however, its efficacy remains controversial due to the limited number of cases, and more information on optimal serum concentrations and appropriate titration methods is required.


Subject(s)
Anticonvulsants/pharmacology , Arrhythmias, Cardiac/chemically induced , Epilepsies, Partial/drug therapy , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Quinidine/pharmacology , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Anticonvulsants/blood , Child , Child, Preschool , Drug Monitoring , Electrocardiography , Female , Humans , Infant , Male , Potassium Channels, Sodium-Activated , Quinidine/administration & dosage , Quinidine/adverse effects , Quinidine/blood
11.
Circulation ; 139(20): 2304-2314, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30696267

ABSTRACT

BACKGROUND: Polymorphic ventricular tachycardia (VT) without QT prolongation is well described in patients without structural heart disease (mainly idiopathic ventricular fibrillation and Brugada syndrome) and in patients with acute ST-elevation myocardial infarction. METHODS: Retrospective study of patients with polymorphic VT related to coronary artery disease, but without evidence of acute myocardial ischemia. RESULTS: The authors identified 43 patients in whom polymorphic VT developed within days of an otherwise uncomplicated myocardial infarction or coronary revascularization procedure. The polymorphic VT events were invariably triggered by extrasystoles with short (364±36 ms) coupling interval. Arrhythmic storms (4-16 events of polymorphic VT deteriorating to ventricular fibrillation) occurred in 23 (53%) patients. These arrhythmic storms were always refractory to conventional antiarrhythmic therapy, including intravenous amiodarone, but invariably responded to quinidine therapy. In-hospital mortality was 17% for patients with arrhythmic storm. Patients treated with quinidine invariably survived to hospital discharge. During long-term follow-up (of 5.6±6 years; range, 1 month to 18 years), 3 (16%) of patients discharged without quinidine developed recurrent polymorphic VT. There were no recurrent arrhythmias during quinidine therapy Conclusions: Arrhythmic storm with recurrent polymorphic VT in patients with coronary disease responds to quinidine therapy when other antiarrhythmic drugs (including intravenous amiodarone) fail.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Coronary Artery Disease/complications , Quinidine/therapeutic use , Tachycardia, Ventricular/drug therapy , Aged , Amiodarone/pharmacology , Amiodarone/therapeutic use , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/pharmacology , Drug Evaluation , Drug Resistance , Drug Substitution , Electrocardiography , Female , Follow-Up Studies , Hospital Mortality , Humans , Male , Middle Aged , Myocardial Infarction/complications , Myocardial Revascularization , Postoperative Complications/drug therapy , Quinidine/adverse effects , Recurrence , Retrospective Studies , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/physiopathology , Thrombocytopenia/chemically induced , Ventricular Fibrillation/etiology , Ventricular Premature Complexes/etiology
12.
PLoS One ; 13(1): e0191514, 2018.
Article in English | MEDLINE | ID: mdl-29352276

ABSTRACT

Non-uniform shortening of the action potential duration (APD90) in different myocardial regions upon heart rate acceleration can set abnormal repolarization gradients and promote arrhythmia. This study examined whether spatial heterogeneities in APD90 restitution can be amplified by drugs with clinically proved proarrhythmic potential (dofetilide, quinidine, procainamide, and flecainide) and, if so, whether these effects can translate to the appropriate changes of the ECG metrics of ventricular repolarization, such as JT intervals. In isolated, perfused guinea-pig heart preparations, monophasic action potentials and volume-conducted ECG were recorded at progressively increased pacing rates. The APD90 measured at distinct ventricular sites, as well as the JTpeak and JTend values were plotted as a function of preceding diastolic interval, and the maximum slopes of the restitution curves were determined at baseline and upon drug administration. Dofetilide, quinidine, and procainamide reverse rate-dependently prolonged APD90 and steepened the restitution curve, with effects being greater at the endocardium than epicardium, and in the right ventricular (RV) vs. the left ventricular (LV) chamber. The restitution slope was increased to a greater extent for the JTend vs. the JTpeak interval. In contrast, flecainide reduced the APD90 restitution slope at LV epicardium without producing effect at LV endocardium and RV epicardium, and reduced the JTpeak restitution slope without changing the JTend restitution. Nevertheless, with all agents, these effects translated to the amplified epicardial-to-endocardial and the LV-to-RV non-uniformities in APD90 restitution, paralleled by the increased JTend vs. JTpeak difference in the restitution slope. In summary, these findings suggest that arrhythmic drug profiles are partly attributable to the accentuated regional heterogeneities in APD90 restitution, which can be indirectly determined through ECG assessments of the JTend vs. JTpeak dynamics at variable pacing rates.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Electrophysiological Phenomena , Female , Flecainide/adverse effects , Guinea Pigs , In Vitro Techniques , Perfusion , Phenethylamines/adverse effects , Procainamide/adverse effects , Quinidine/adverse effects , Sulfonamides/adverse effects
13.
Neurology ; 90(1): e67-e72, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29196578

ABSTRACT

OBJECTIVE: To evaluate quinidine as a precision therapy for severe epilepsy due to gain of function mutations in the potassium channel gene KCNT1. METHODS: A single-center, inpatient, order-randomized, blinded, placebo-controlled, crossover trial of oral quinidine included 6 patients with severe autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE) due to KCNT1 mutation. Order was block randomized and blinded. Four-day treatment blocks were used with a 2-day washout between. Dose started at 900 mg over 3 divided doses then, in subsequent participants, was reduced to 600 mg, then 300 mg. Primary outcome was seizure frequency measured on continuous video-EEG in those completing the trial. RESULTS: Prolonged QT interval occurred in the first 2 patients at doses of 900 and 600 mg quinidine per day, respectively, despite serum quinidine levels well below the therapeutic range (0.61 and 0.51 µg/mL, reference range 1.3-5.0 µg/mL). Four patients completed treatment with 300 mg/d without adverse events. Patients completing the trial had very frequent seizures (mean 14 per day, SD 7, median 13, interquartile range 10-18). Seizures per day were nonsignificantly increased by quinidine (median 2, 95% confidence interval -1.5 to +5, p = 0.15) and no patient had a 50% seizure reduction. CONCLUSION: Quinidine did not show efficacy in adults and teenagers with ADNFLE. Dose-limiting cardiac side effects were observed even in the presence of low measured serum quinidine levels. Although small, this trial suggests use of quinidine in ADNFLE is likely to be ineffective coupled with considerable cardiac risks. CLINICAL TRIALS REGISTRATION: Australian Therapeutic Goods Administration Clinical Trial Registry (trial number 2015/0151). CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that for persons with severe epilepsy due to gain of function mutations in the potassium channel gene KCNT1, quinidine does not significantly reduce seizure frequency.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy, Frontal Lobe/drug therapy , Epilepsy, Frontal Lobe/genetics , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Precision Medicine , Quinidine/therapeutic use , Adolescent , Adult , Anticonvulsants/adverse effects , Anticonvulsants/blood , Cross-Over Studies , Double-Blind Method , Epilepsy, Frontal Lobe/blood , Gain of Function Mutation , Humans , Middle Aged , Potassium Channels, Sodium-Activated , Quinidine/adverse effects , Quinidine/blood , Seizures/blood , Seizures/drug therapy , Seizures/genetics , Treatment Failure
14.
Mini Rev Med Chem ; 18(6): 468-475, 2018.
Article in English | MEDLINE | ID: mdl-28685701

ABSTRACT

Historically, quinidine was the first medicine used in the therapy of heart arrhythmias. Studies in the early 20th century identified quinidine, a diastereomer of the antimalarial quinine, as the most potent of the antiarrhythmic substances extracted from the cinchona plant. Quinidine is used by the 1920s, as an antiarrhythmic agent to maintain sinus rhythm after the conversion from atrial flutter or atrial fibrillation and to prevent recurrence of ventricular tachycardia or ventricular fibrillation. Its value in chronic prophylaxis of relapse of ventricular arrhythmia was brought under suspicion after publishing of meta analysis that showed that the application of quinidine increases mortality. Due to numerous proofs of increased risk for the appearance of ventricular arrhythmia and sudden death, as well as a number of other adverse effects and drug interactions, quinidine was withdrawn from use and in the recent years has become unavailable in many countries. On the other hand, recent studies have demonstrated that quinidine is the only oral medication that has consistently shown efficacy in preventing arrhythmias and terminating storms due to recurrent ventricular fibrillation, in patients with Brugada syndrome, idiopathic ventricular fibrillation and early repolarization syndrome. Quinidine is also the only antiarrhythmic drug that normalized the QT interval in patients with the congenital short QT syndrome. The aim of this review is to provide good insight into pro and contra arguments for quinidine use in ventricular arrhythmias evidence based on recently published literature.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Quinidine/therapeutic use , Ventricular Fibrillation/drug therapy , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Brugada Syndrome/drug therapy , Humans , Quinidine/adverse effects , Quinidine/pharmacokinetics , Quinidine/pharmacology
17.
Mov Disord ; 32(6): 893-903, 2017 06.
Article in English | MEDLINE | ID: mdl-28370447

ABSTRACT

BACKGROUND: Nondopaminergic pathways represent potential targets to treat levodopa-induced dyskinesia in Parkinson's disease (PD). This pilot-study (NCT01767129) examined the safety/efficacy of the sigma-1 receptor-agonist and glutamatergic/monoaminergic modulator, dextromethorphan plus quinidine (to inhibit rapid dextromethorphan metabolism), for treating levodopa-induced dyskinesia. METHODS: PD patients were randomized to dextromethorphan/quinidine (45 mg/10 mg twice daily)/placebo in two 2-week double-blind, crossover treatment periods, with intervening 2-week washout. After 14 days, a 2-hour intravenous levodopa-infusion was administered. Patient examinations were videotaped before infusion ("off" state) and every 30 minutes during and afterwards until patients returned to "off." The primary endpoint was dyskinesia-severity during infusion measured by Unified Dyskinesia Rating Scale part 3 area-under-curve scores (blinded expert rated). Additional endpoints included other dyskinesia/motor assessments, global measures of clinical-change, and adverse-events. RESULTS: A total of 13 patients were randomized and completed the study (efficacy-evaluable population). Dyskinesia-severity was nonsignificantly lower with dextromethorphan/quinidine than placebo during infusion (area-under-curve 966.5 vs 1048.8; P = .191 [efficacy-evaluable patients]), and significantly lower in a post-hoc sensitivity analysis of the per-protocol-population (efficacy-evaluable patients with ≥ 80% study-drug-compliance, n = 12) when measured from infusion start to 4-hours post-infusion completion (area-under-curve 1585.0 vs 1911.3; P = .024). Mean peak dyskinesia decreased significantly from infusion-start to return to "off" (13.3 vs 14.9; P = .018 [efficacy-evaluable patients]). A total of 9 patients rated dyskinesia "much/very much improved" on dextromethorphan/quinidine versus 1-patient on placebo. Dextromethorphan/quinidine did not worsen PD-motor scores, was generally well tolerated, and was associated with more frequent adverse events. CONCLUSION: This study provides preliminary evidence of clinical benefit with dextromethorphan/quinidine for treating levodopa-induced dyskinesia in PD. Larger studies with a longer treatment duration need to corroborate these early findings. © 2017 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dextromethorphan/pharmacology , Dyskinesia, Drug-Induced/drug therapy , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Levodopa/adverse effects , Outcome Assessment, Health Care , Parkinson Disease/drug therapy , Quinidine/pharmacology , Aged , Cross-Over Studies , Dextromethorphan/administration & dosage , Dextromethorphan/adverse effects , Double-Blind Method , Drug Therapy, Combination , Dyskinesia, Drug-Induced/etiology , Enzyme Inhibitors/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/adverse effects , Female , Humans , Male , Middle Aged , Pilot Projects , Quinidine/administration & dosage , Quinidine/adverse effects
18.
Mol Biosyst ; 13(5): 1018-1030, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28418441

ABSTRACT

Wenxin Keli (WK), a Chinese patent medicine, is known to be effective against cardiac arrhythmias and heart failure. Although a number of electrophysiological findings regarding its therapeutic effect have been reported, the active components and system-level characterizations of the component-target interactions of WK have yet to be elucidated. In the current study, we present the first report of a new protective effect of WK on suppressing anti-arrhythmic-agent-induced arrhythmias. In a model of isolated guinea pig hearts, rapid perfusion of quinidine altered the heart rate and prolonged the Q-T interval. Pretreatment with WK significantly prevented quinidine-induced arrhythmias. To explain the therapeutic and protective effects of WK, we constructed an integrated multi-target pharmacological mechanism prediction workflow in combination with machine learning and molecular pathway analysis. This workflow had the ability to predict and rank the probability of each compound interacting with 1715 target proteins simultaneously. The ROC value statistics showed that 97.786% of the values for target prediction were larger than 0.8. We applied this model to carry out target prediction and network analysis for the identified components of 5 herbs in WK. Using the 124 potential anti-arrhythmic components and the 30 corresponding protein targets obtained, an integrative anti-arrhythmic molecular mechanism of WK was proposed. Emerging drug/target networks suggested ion channel and intracellular calcium and autonomic nervous and hormonal regulation had critical roles in WK-mediated anti-arrhythmic activity. A validation of the proposed mechanisms was achieved by demonstrating that calaxin, one of the WK components from Gansong, dose-dependently blocked its predicted target CaV1.2 channel in an electrophysiological assay.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Gene Regulatory Networks/drug effects , Heart/drug effects , Animals , Calcium Channels, L-Type/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Heart/physiopathology , Machine Learning , Models, Biological , Quinidine/adverse effects , ROC Curve , Signal Transduction
19.
Stem Cell Res Ther ; 8(1): 54, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28279214

ABSTRACT

BACKGROUND: Cardiotoxicity remains an important concern in drug discovery. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have become an attractive platform to evaluate cardiotoxicity. However, the consistency between human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in prediction of cardiotoxicity has yet to be elucidated. METHODS: Here we screened the toxicities of four representative drugs (E-4031, isoprenaline, quinidine, and haloperidol) using both hESC-CMs and hiPSC-CMs, combined with an impedance-based bioanalytical method. RESULTS: It showed that both hESC-CMs and hiPSC-CMs can recapitulate cardiotoxicity and identify the effects of well-characterized compounds. CONCLUSIONS: The combined platform of hPSC-CMs and an impedance-based bioanalytical method could improve preclinical cardiotoxicity screening, holding great potential for increasing drug development accuracy.


Subject(s)
Cardiotoxicity/physiopathology , Human Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Cell Differentiation/genetics , Drug Discovery , Haloperidol/adverse effects , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Isoproterenol/adverse effects , Piperidines/adverse effects , Pyridines/adverse effects , Quinidine/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...