Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
Add more filters

Publication year range
1.
J Electrocardiol ; 69S: 55-60, 2021.
Article in English | MEDLINE | ID: mdl-34736759

ABSTRACT

INTRODUCTION: Drug-induced block of the hERG potassium channel could predispose to torsade de pointes, depending on occurrence of concomitant blocks of the calcium and/or sodium channels. Since the hERG potassium channel block affects cardiac repolarization, the aim of this study was to propose a new reliable index for non-invasive assessment of drug-induced hERG potassium channel block based on electrocardiographic T-wave features. METHODS: ERD30% (early repolarization duration) and TS/A (down-going T-wave slope to T-wave amplitude ratio) features were measured in 22 healthy subjects who received, in different days, doses of dofetilide, ranolazine, verapamil and quinidine (all being hERG potassium channel blockers and the latter three being also blockers of calcium and/or sodium channels) while undergoing continuous electrocardiographic acquisition from which ERD30% and TS/A were evaluated in fifteen time points during the 24 h following drug administration ("ECG Effects of Ranolazine, Dofetilide, Verapamil, and Quinidine in Healthy Subjects" database by Physionet). A total of 1320 pairs of ERD30% and TS/A measurements, divided in training (50%) and testing (50%) datasets, were obtained. Drug-induced hERG potassium channel block was modelled by the regression equation BECG(%) = a·ERD30% + b·TS/A+ c·ERD30%·TS/A + d; BECG(%) values were compared to plasma-based measurements, BREF(%). RESULTS: Regression coefficients values, obtained on the training dataset, were: a = -561.0 s-1, b = -9.7 s, c = 77.2 and d = 138.9. In the testing dataset, correlation coefficient between BECG(%) and BREF(%) was 0.67 (p < 10-81); estimation error was -11.5 ± 16.7%. CONCLUSION: BECG(%) is a reliable non-invasive index for the assessment of drug-induced hERG potassium channel block, independently from concomitant blocks of other ions.


Subject(s)
Electrocardiography , Pharmaceutical Preparations , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Potassium Channel Blockers/adverse effects , Verapamil
2.
Laeknabladid ; 107(4): 179-184, 2021 Apr.
Article in Is | MEDLINE | ID: mdl-33769308

ABSTRACT

INTRODUCTION: Fampridine is a drug for people with Multiple Sclerosis (MS). It is a broad-spectrum voltage-dependent potassium channel blocker that enhances synaptic transmission. The drug has been shown to be able to enhance conduction in demyelinated axons, thereby leading to improved gait in patients with MS. The purpose of this study was to examine the effect of fampridine on gait function in people with MS in the end of a 2 weeks trial drug period and to observe how many patients continued drug therapy. MATERIAL AND METHODS: Data from 41 individuals with MS was collected retrospectively for this study. Measurements were administered by physiotherapists and the results from the Timed 25-Foot Walk (T25FW) and 12-item Multiple Sclerosis Walking Scale (MSWS-12) were obtained from medical records from The National University Hospital of Iceland. RESULTS: The results showed a significant difference in walking speed before and at the end of trial period (p<0.0001). The average improvement in walking speed was 22%. Results also demonstrated a significant difference in MSWS-12 scores before and at the end of treatment (p<0.0001). The average improvement in MSWS-12 was 11.4 points. Eighteen individuals (43.9%) continued treatment after the trial period. CONCLUSION: Fampridine can have a positive effect on impaired gait function in people with MS and can be an important adjunct to treatment.


Subject(s)
Multiple Sclerosis , 4-Aminopyridine/adverse effects , Gait , Humans , Multiple Sclerosis/diagnosis , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/adverse effects , Retrospective Studies , Walking
3.
Mol Pharmacol ; 95(5): 537-550, 2019 05.
Article in English | MEDLINE | ID: mdl-30770456

ABSTRACT

Current guidelines around preclinical screening for drug-induced arrhythmias require the measurement of the potency of block of voltage-gated potassium channel subtype 11.1 (Kv11.1) as a surrogate for risk. A shortcoming of this approach is that the measured IC50 of Kv11.1 block varies widely depending on the voltage protocol used in electrophysiological assays. In this study, we aimed to investigate the factors that contribute to these differences and to identify whether it is possible to make predictions about protocol-dependent block that might facilitate the comparison of potencies measured using different assays. Our data demonstrate that state preferential binding, together with drug-binding kinetics and trapping, is an important determinant of the protocol dependence of Kv11.1 block. We show for the first time that differences in IC50 measured between protocols occurs in a predictable way, such that machine-learning algorithms trained using a selection of simple voltage protocols can indeed predict protocol-dependent potency. Furthermore, we also show that the preference of a drug for binding to the open versus the inactivated state of Kv11.1 can also be inferred from differences in IC50 values measured between protocols. Our work therefore identifies how state preferential drug binding is a major determinant of the protocol dependence of IC50 values measured in preclinical Kv11.1 assays. It also provides a novel method for quantifying the state dependence of Kv11.1 drug binding that will facilitate the development of more complete models of drug binding to Kv11.1 and improve our understanding of proarrhythmic risk associated with compounds that block Kv11.1.


Subject(s)
Biological Assay/methods , ERG1 Potassium Channel/genetics , ERG1 Potassium Channel/metabolism , Potassium Channel Blockers/pharmacology , Animals , Arrhythmias, Cardiac/chemically induced , CHO Cells , Cell Line , Cricetulus , Electrophysiological Phenomena/drug effects , Humans , Inhibitory Concentration 50 , Kinetics , Potassium Channel Blockers/adverse effects
4.
BMC Neurosci ; 20(1): 62, 2019 12 21.
Article in English | MEDLINE | ID: mdl-31864286

ABSTRACT

BACKGROUND: Sodium ion transportation plays a crucial role in the pathogenesis of hypoxic-ischemic brain injury. Amiodarone, a Vaughan-Williams class III antiarrhythmic drug, has been widely used to treat life-threatening arrhythmia and cardiac arrest worldwide. In addition to its inhibitory effects on the potassium channel, amiodarone also blocks various sodium ion transporters, including the voltage-gated sodium channel, sodium pump, and Na+/Ca+ exchanger. Considering these pharmacological profile, amiodarone may affect the influx-efflux balance of sodium ion in the hypoxic-ischemic brain. Previous studies suggest that the blockade of the voltage-gated sodium channel during hypoxic-ischemic brain injury exerts neuroprotection. On the contrary, the blockade of sodium pump or Na+/Ca+ exchanger during hypoxia-ischemia may cause further intracellular sodium accumulation and consequent osmotic cell death. From these perspectives, the effects of amiodarone on sodium ion balance on the hypoxic-ischemic brain can be both protective and detrimental depending on the clinical and pathophysiological conditions. In this study, we therefore investigated the effect of amiodarone on hypoxic-ischemic brain injury using a murine experimental model. RESULTS: Compared with the control group mice, mice that received amiodarone after induction of 40-min hypoxic-ischemic brain injury exhibited lower survival rates over 7 days and worse neurological function. After 25-min hypoxic-ischemic brain injury, amiodarone treated mice exhibited larger infarct volumes (16.0 ± 6.9 vs. 24.2 ± 6.8 mm3, P < 0.05) and worse neurological function. In addition, the brains harvested from the amiodarone-treated mice contained larger amounts of sodium (194.7 ± 45.1 vs. 253.5 ± 50.9 mEq/kg dry weight, P < 0.01) and water (259.3 ± 8.9 vs. 277.2 ± 12.5 mg, P < 0.01). There were no significant differences in hemodynamic parameters between groups. CONCLUSIONS: Amiodarone exacerbated brain injuries and neurological outcomes after hypoxic-ischemic insults. Severe brain sodium accumulation and brain edema were associated with the detrimental effects of amiodarone. Amiodarone at the clinical dose can exacerbate brain injury after hypoxic-ischemic insult by affecting sodium ion transportation and facilitate intracellular sodium accumulation in the brain.


Subject(s)
Amiodarone/adverse effects , Brain/drug effects , Brain/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Sodium Channel Blockers/adverse effects , Animals , Anti-Arrhythmia Agents/adverse effects , Brain/pathology , Brain Edema/pathology , Brain Edema/physiopathology , Disease Models, Animal , Hypoxia-Ischemia, Brain/pathology , Male , Mice, Inbred C57BL , Neuroprotection/drug effects , Potassium Channel Blockers/adverse effects , Sodium/metabolism
5.
Mult Scler ; 25(4): 618-621, 2019 04.
Article in English | MEDLINE | ID: mdl-30375922

ABSTRACT

BACKGROUND: Dalfampridine extended release (DAL) is a broad-spectrum voltage-gated potassium channel blocker that is indicated in multiple sclerosis to improve the nerve conduction of demyelinated axons. Seizures are a known side effect of DAL, which is contraindicated in patients with a history of epilepsy. OBJECTIVE: Three cases of multiple sclerosis (MS) with de novo convulsive status epilepticus (CSE) probably related to dalfampridine administration are described. METHODS: No patients had a history of seizures or renal impairment. Biological tests were normal. A brain magnetic resonance imaging (MRI) showed diffuse cortical and subcortical atrophy without active inflammatory lesions. RESULTS: All three patients presented with CSE that was attributed to DAL and so was discontinued. CONCLUSION: These case reports illustrate that, aside from seizures, de novo CSE is a potential complication of MS patients treated with DAL.


Subject(s)
4-Aminopyridine/adverse effects , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/adverse effects , Status Epilepticus/chemically induced , Adult , Female , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy
6.
Med Res Rev ; 38(2): 525-555, 2018 03.
Article in English | MEDLINE | ID: mdl-28467598

ABSTRACT

Drug-induced blockade of human ether-a-go-go-related gene (hERG) remains a major impediment in delivering safe drugs to the market. Several drugs have been withdrawn from the market due to their severe cardiotoxic side effects triggered by their off-target interactions with hERG. Thus, identifying the potential hERG blockers at early stages of lead discovery is fast evolving as a standard in drug design and development. A number of in silico structure-based models of hERG have been developed as a low-cost solution to evaluate drugs for hERG liability, and it is now agreed that the hERG blockers bind at the large central cavity of the channel. Nevertheless, there is no clear convergence on the appropriate drug binding modes against the channel. The proposed binding modes differ in their orientations and interpretations on the role of key residues in the channel. Such ambiguities in the modes of binding remain to be a significant challenge in achieving efficient computational predictive models and in saving many important already Food and Drug Administration approved drugs. In this review, we discuss the spectrum of reported binding modes for hERG blockers, the various in silico models developed for predicting a drug's affinity to hERG, and the known successful optimization strategies to avoid off-target interactions with hERG.


Subject(s)
Drug Discovery , Ether-A-Go-Go Potassium Channels/metabolism , Potassium Channel Blockers/adverse effects , Binding Sites , Cardiotoxicity/pathology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/chemistry , Humans , Potassium Channel Blockers/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
7.
Acta Pharmacol Sin ; 39(5): 683-694, 2018 May.
Article in English | MEDLINE | ID: mdl-29671418

ABSTRACT

ATP-sensitive potassium (KATP) channels are ubiquitously expressed on the plasma membrane of cells in multiple organs, including the heart, pancreas and brain. KATP channels play important roles in controlling and regulating cellular functions in response to metabolic state, which are inhibited by ATP and activated by Mg-ADP, allowing the cell to couple cellular metabolic state (ATP/ADP ratio) to electrical activity of the cell membrane. KATP channels mediate insulin secretion in pancreatic islet beta cells, and controlling vascular tone. Under pathophysiological conditions, KATP channels play cytoprotective role in cardiac myocytes and neurons during ischemia and/or hypoxia. KATP channel is a hetero-octameric complex, consisting of four pore-forming Kir6.x and four regulatory sulfonylurea receptor SURx subunits. These subunits are differentially expressed in various cell types, thus determining the sensitivity of the cells to specific channel modifiers. Sulfonylurea class of antidiabetic drugs blocks KATP channels, which are neuroprotective in stroke, can be one of the high stoke risk factors for diabetic patients. In this review, we discussed the potential effects of KATP channel blockers when used under pathological conditions related to diabetics and cerebral ischemic stroke.


Subject(s)
Cerebral Infarction/prevention & control , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/therapeutic use , KATP Channels/metabolism , Potassium Channel Blockers/therapeutic use , Animals , Cerebral Infarction/physiopathology , Diabetes Mellitus/physiopathology , Humans , Hypoglycemic Agents/adverse effects , KATP Channels/physiology , Potassium Channel Blockers/adverse effects , Sulfonylurea Compounds/adverse effects , Sulfonylurea Compounds/therapeutic use
8.
J Pharmacol Sci ; 134(2): 75-85, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28615142

ABSTRACT

Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes hold great potentials to predict pro-arrhythmic risks in preclinical cardiac safety screening, although the hiPSC cardiomyocytes exhibit rather immature functional and structural characteristics, including spontaneous activity. Our physiological characterization and mathematical simulation showed that low expression of the inward-rectifier potassium (IK1) channel is a determinant of spontaneous activity. To understand impact of the low IK1 expression on the pharmacological properties, we tested if transduction of hiPSC-derived cardiomyocytes with KCNJ2, which encodes the IK1 channel, alters pharmacological response to cardiac repolarization processes. The transduction of KCNJ2 resulted in quiescent hiPSC-derived cardiomyocytes, which need pacing to elicit action potentials. Significant prolongation of paced action potential duration in KCNJ2-transduced hiPSC-derived cardiomyocytes was stably measured at 0.1 µM E-4031, although the same concentration of E-4031 ablated firing of non-treated hiPSC-derived cardiomyocytes. These results in single cells were confirmed by mathematical simulations. Using the hiPSC-derived cardiac sheets with KCNJ2-transduction, we also investigated effects of a range of drugs on field potential duration recorded at 1 Hz. The KCNJ2 overexpression in hiPSC-derived cardiomyocytes may contribute to evaluate a part of QT-prolonging drugs at toxicological concentrations with high accuracy.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Potassium Channel Blockers/adverse effects , Potassium Channels, Inwardly Rectifying/metabolism , Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Drug Evaluation, Preclinical/methods , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Piperidines/adverse effects , Pyridines/adverse effects
9.
Arch Toxicol ; 91(12): 3885-3895, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28551711

ABSTRACT

Recent reports have noted that a number of compounds that block the human Ether-à-go-go related gene (hERG) ion channel also induce phospholipidosis (PLD). To explore a hypothesis explaining why most PLD inducers are also hERG inhibitors, a modeling approach was undertaken with data sets comprised of 4096 compounds assayed for hERG inhibition and 5490 compounds assayed for PLD induction. To eliminate the chemical domain effect, a filtered data set of 567 compounds tested in quantitative high-throughput screening (qHTS) format for both hERG inhibition and PLD induction was constructed. Partial least squares (PLS) modeling followed by 3D-SDAR mapping of the most frequently occurring bins and projection on to the chemical structure suggested that both adverse effects are driven by similar structural features, namely two aromatic rings and an amino group forming a three-center toxicophore. Non-parametric U-tests performed on the original 3D-SDAR bins indicated that the distance between the two aromatic rings is the main factor determining the differences in activity; at distances of up to about 5.5 Å, a phospholipidotic compound would also inhibit hERG, while at longer distances, a sharp reduction of the PLD-inducing potential leaves only a well-pronounced hERG blocking effect. The hERG activity itself diminishes after the distance between the centroids of the two aromatic rings exceeds 12.5 Å. Further comparison of the two toxicophores revealed that the almost identical aromatic rings to amino group distances play no significant role in distinguishing between PLD and hERG activity. The hypothesis that the PLD toxicophore appears to be a subset of the hERG toxicophore explains why about 80% of all phospholipidotic chemicals (the remaining 20% are thought to act via a different mechanism) also inhibit the hERG ion channel. These models were further validated in large-scale qHTS assays testing 1085 chemicals for their PLD-inducing potential and 1570 compounds for hERG inhibition. After removal of the modeling and experimental inconclusive compounds, the area under the receiver-operating characteristic (ROC) curve was 0.92 for the PLD model and 0.87 for the hERG model. Due to the exceptional ability of these models to recognize safe compounds (negative predictive values of 0.99 for PLD and 0.94 for hERG were achieved), their use in regulatory settings might be particularly useful.


Subject(s)
ERG1 Potassium Channel/antagonists & inhibitors , Lipidoses/chemically induced , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Quantitative Structure-Activity Relationship , Algorithms , Humans , Models, Molecular , Phospholipids/metabolism , Potassium Channel Blockers/adverse effects , Reproducibility of Results
10.
J Cardiovasc Electrophysiol ; 27(10): 1206-1213, 2016 10.
Article in English | MEDLINE | ID: mdl-27333947

ABSTRACT

BACKGROUND: Efavirenz (EFV) has been associated with torsade de pointes despite marginal QT interval lengthening. Since EFV is metabolized by the cytochrome P450 (CYP) 2B6 enzyme, we hypothesized that EFV would lengthen the rate-corrected QT (QTcF) interval in carriers of the CYP2B6*6 decreased functional allele. OBJECTIVE: The primary objective of this study was to evaluate EFV-associated QT interval changes with regard to CYP2B6 genotype and to explore mechanisms of QT interval lengthening. METHODS: EFV was administered to healthy volunteers (n = 57) as a single 600 mg dose followed by multiple doses to steady-state. Subjects were genotyped for known CYP2B6 alleles and ECGs and EFV plasma concentrations were obtained serially. Whole-cell, voltage-clamp experiments were performed on cells stably expressing hERG and exposed to EFV in the presence and absence of CYP2B6 expression. RESULTS: EFV demonstrated a gene-dose effect and exceeded the FDA criteria for QTcF interval prolongation in CYP2B6*6/*6 carriers. The largest mean time-matched differences ∆∆QTcF were observed at 6 hours (14 milliseconds; 95% CI [1; 27]), 12 hours (18 milliseconds; 95% CI [-4; 40]), and 18 hours (6 milliseconds; 95% CI [-1; 14]) in the CYP2B6*6/*6 genotype. EFV concentrations exceeding 0.4 µg/mL significantly inhibited outward hERG tail currents (P < 0.05). CONCLUSIONS: This study demonstrates that homozygous carriers of CYP2B6*6 allele may be at increased risk for EFV-induced QTcF interval prolongation via inhibition of hERG.


Subject(s)
Benzoxazines/adverse effects , Cytochrome P-450 CYP2B6/genetics , ERG1 Potassium Channel/antagonists & inhibitors , Pharmacogenomic Variants , Potassium Channel Blockers/adverse effects , Reverse Transcriptase Inhibitors/adverse effects , Torsades de Pointes/chemically induced , Action Potentials , Adolescent , Adult , Alkynes , Benzoxazines/blood , Cyclopropanes , Cytochrome P-450 CYP2B6/metabolism , Dose-Response Relationship, Drug , ERG1 Potassium Channel/metabolism , Electrocardiography , Female , Gene Frequency , Genotype , HEK293 Cells , Healthy Volunteers , Heart Rate/drug effects , Homozygote , Humans , Male , Pharmacogenetics , Phenotype , Potassium Channel Blockers/blood , Reverse Transcriptase Inhibitors/blood , Risk Assessment , Risk Factors , Time Factors , Torsades de Pointes/genetics , Torsades de Pointes/metabolism , Torsades de Pointes/physiopathology , Transfection , Young Adult
11.
Mult Scler ; 22(7): 944-54, 2016 06.
Article in English | MEDLINE | ID: mdl-26447066

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is a debilitating disease that negatively impacts patients' lives. OBJECTIVE: ENABLE assessed the effect of long-term prolonged-release (PR) fampridine (dalfampridine extended release in the United States) treatment on patient-perceived health impact in patients with MS with walking impairment. METHODS: ENABLE was a 48-week, open-label, Phase 4 study of PR-fampridine 10 mg twice daily. Patients who showed any improvement in Timed 25-Foot Walk walking speed at weeks 2 and 4 and any improvement in 12-item MS Walking Scale score at week 4 remained on treatment. The primary endpoint was change from baseline in 36-Item Short-Form Health Survey (SF-36) physical component summary (PCS) score. RESULTS: At week 4, 707/901 (78.5%) patients met the criteria to remain on treatment. Patients on treatment demonstrated significant and clinically meaningful improvements in SF-36 PCS scores from baseline (mean change (95% confidence interval)) to week 12 (4.30 (3.83, 4.78); p < 0.0001), week 24 (3.75 (3.23, 4.27); p < 0.0001), week 36 (3.46 (2.95, 3.97); p < 0.0001), and week 48 (3.24 (2.72, 3.77); p < 0.0001). Significant improvements from baseline were also demonstrated in secondary health measures in patients on treatment. CONCLUSION: PR-fampridine improved patient-perceived physical and psychological health impact of MS measured in a real-life setting.


Subject(s)
4-Aminopyridine/therapeutic use , Multiple Sclerosis/drug therapy , Patient Reported Outcome Measures , Potassium Channel Blockers/therapeutic use , 4-Aminopyridine/adverse effects , Adolescent , Adult , Aged , Australia , Delayed-Action Preparations , Disability Evaluation , Europe , Exercise Tolerance/drug effects , Female , Gait , Humans , Male , Middle Aged , Multiple Sclerosis/diagnosis , Multiple Sclerosis/physiopathology , Multiple Sclerosis/psychology , Potassium Channel Blockers/adverse effects , Quality of Life , Recovery of Function , Time Factors , Treatment Outcome , Walking , Young Adult
12.
Europace ; 18(3): 340-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26462707

ABSTRACT

AIMS: To assess the efficacy of BMS 914392 on atrial fibrillation (AF) burden reduction in 20 patients with pacemakers and paroxysmal atrial fibrillation (PAF). BMS 914392 is a potent, selective, oral inhibitor of the IKACh current and has been shown to suppress AF, whilst having no effect on the ventricular refractory period. This is the first efficacy study of BMS 914392 in patients with PAF. METHODS AND RESULTS: The study was a four-way, crossover, double-blind design. A total of 20 patients with PAF and dual-chamber pacemakers were recruited. The pacemakers allowed beat-to-beat monitoring. Anti-arrhythmic drugs were withdrawn. Patients received low-dose (10 mg OD), medium-dose (10 mg TDS), and high-dose (20 mg TDS) BMS 914392 or placebo for 3 weeks before being crossed to the next phase. Patients underwent a washout period, four treatment phases and a final washout phase. Atrial fibrillation burden was downloaded from their pacemakers at the end of each study phase. BMS 914392 did not reduce AF burden when compared with placebo (10 mg OD P = 0.56, 10 mg TDS P = 0.22, 20 mg TDS P = 0.23). Heart rate and corrected QT (QTc) were not affected by BMS 914392. Adverse event (AE) rates did not differ from placebo in any of the treatment groups, with no serious AEs recorded. CONCLUSION: BMS 914932 has not been shown to reduce AF burden in patients with PAF and pacemakers using beat-to-beat pacemaker monitoring throughout the study. BMS 914392 was well tolerated and did not affect QTc or reduce heart rate. TRIAL REGISTRATION: Clinicaltrials.gov: NCT01356914.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/therapy , Cardiac Resynchronization Therapy Devices , Cardiac Resynchronization Therapy , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , Heart Conduction System/drug effects , Heart Rate/drug effects , Potassium Channel Blockers/therapeutic use , Pyrans/therapeutic use , Quinolines/therapeutic use , Action Potentials , Aged , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Cross-Over Studies , Double-Blind Method , Electrocardiography , England , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Humans , Male , Potassium Channel Blockers/adverse effects , Pyrans/adverse effects , Quinolines/adverse effects , Time Factors , Treatment Outcome
13.
Yao Xue Xue Bao ; 51(10): 1530-9, 2016 10.
Article in Zh | MEDLINE | ID: mdl-29932317

ABSTRACT

The potassium channel encoded by the human ether-a-go-go related gene(hERG) plays a very important role in the physiological and pathological processes in human. hERG potassium channel determines the outward currents which facilitate the repolarization of the myocardial cells. Some drugs were withdrawn from the market for the serious side effect of long QT interval and arrhythmia due to blockade of hERG channel. The strategies for lead compound optimization are to reduce inhibitory activity of hERG potassium channel and decrease cardiac toxicity. These methods include reduction of lipophilicity and basicity of amines, introduction of hydroxyl and acidic groups, and restricting conformation.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiotoxicity/prevention & control , Ether-A-Go-Go Potassium Channels/physiology , Long QT Syndrome/chemically induced , Myocytes, Cardiac/physiology , Potassium Channel Blockers/adverse effects , Drug-Related Side Effects and Adverse Reactions , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Humans
14.
J Cardiovasc Pharmacol ; 66(5): 412-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25978691

ABSTRACT

There is a clear unmet medical need for new pharmacologic therapies with improved efficacy and safety for the treatment of atrial fibrillation. Considerable research efforts have been undertaken to discover and develop new safe and effective antiarrhythmic drugs that specifically target atrial K(+) channels. To realize the full value of these novel atrial-specific therapeutic drug targets, demonstration of clinical efficacy and safety is required for a new breed of atrial-selective antiarrhythmic drugs. The reward for demonstrating this in a pivotal phase III trial, on regulatory approval, will be "first-in-class" status. This article reviews the development status of new and novel K channel inhibitors currently in drug development as atrial-selective antiarrhythmics for the treatment of atrial fibrillation.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Drug Discovery , Drugs, Investigational/therapeutic use , Heart Conduction System/drug effects , Heart Rate/drug effects , Potassium Channel Blockers/therapeutic use , Potassium Channels/drug effects , Action Potentials , Animals , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Drugs, Investigational/adverse effects , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Humans , Potassium Channel Blockers/adverse effects , Potassium Channels/metabolism , Signal Transduction/drug effects
15.
Biol Pharm Bull ; 38(5): 781-4, 2015.
Article in English | MEDLINE | ID: mdl-25947924

ABSTRACT

The human ether-à-go-go-related gene (hERG) channel mediates the rapid delayed rectifier potassium current (IKr) responsible for shaping the repolarization phase of cardiac action potentials. hERG mutation may cause hERG channel malfunction, leading to long QT syndrome and other arrhythmic disorders. Elucidation of the genotype-phenotype relationships of individual hERG mutations is key to the development of treatment for such arrhythmic disorders. We previously identified hERG(G487R), a missense mutant with a glycine-to-arginine substitution at position 487. In the absence of arrhythmogenic factors, hERG(G487R) subunit-containing channels show normal surface expression and gating kinetics. However, it remains unknown whether the mutation exacerbates hERG channel malfunction induced by arrhythmogenic factors. Here we used a voltage-clamp technique to compare the effects of the major arrythmogenic factors on wild-type hERG [hERG(WT)] and hERG(G487R) channel currents (IhERG) in HEK-293T cells. The extent of IhERG blockade by the antiarrhythmic drug dofetilide or E4031 was not different between these channels. On the other hand, the extracellular K(+) concentration ([K(+)]ex)-dependent changes in the rates of recovery from inactivation and deactivation of IhERG were rather less obvious for hERG(G487R) channel than for hERG(WT) channel. These findings suggest that the inheritance of hERG(G487R) does not increase the risk of arrhythmic disorders induced by antiarrhythmic drugs or hypokalemia.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/genetics , Ether-A-Go-Go Potassium Channels/genetics , Heart Conduction System/abnormalities , Mutation , Phenethylamines/adverse effects , Potassium Channel Blockers/adverse effects , Potassium/metabolism , Sulfonamides/adverse effects , Action Potentials/drug effects , Arrhythmias, Cardiac/etiology , Brugada Syndrome , Cardiac Conduction System Disease , Genotype , HEK293 Cells , Heart/drug effects , Heart/physiopathology , Humans , Hypokalemia/complications , Ion Channel Gating/genetics , Kinetics , Long QT Syndrome/etiology , Long QT Syndrome/genetics , Patch-Clamp Techniques , Phenotype
16.
J Electrocardiol ; 48(6): 1081-7, 2015.
Article in English | MEDLINE | ID: mdl-26324176

ABSTRACT

INTRODUCTION: Heart rate corrected QT (QTc) interval prolongation is a predictor of drug-induced torsade de pointes, a potentially fatal ventricular arrhythmia that disproportionately affects women. This study assesses whether there are sex differences in the ECG changes induced by four different hERG potassium channel blocking drugs. METHODS AND RESULTS: Twenty-two healthy subjects (11 women) received a single oral dose of dofetilide, quinidine, ranolazine, verapamil and placebo in a double-blind 5-period crossover study. ECGs and plasma drug concentrations were obtained at pre-dose and at 15 time-points post-dose. Dofetilide, quinidine and ranolazine prolonged QTc. There were no sex differences in QTc prolongation for any drug, after accounting for differences in exposure. Sex differences in any ECG biomarker were observed only with dofetilide, which caused greater J-Tpeakc prolongation (p=0.045) but lesser Tpeak-Tend prolongation (p=0.006) and lesser decrease of T wave amplitude (p=0.003) in women compared to men. CONCLUSIONS: There were no sex differences in QTc prolongation for any of the studied drugs. Moreover, no systematic sex differences in other drug-induced ECG biomarker changes were observed in this study. This study suggests that the higher torsade risk in women compared to men is not due to a larger concentration-dependent QTc prolongation.


Subject(s)
Electrocardiography/drug effects , Potassium Channel Blockers/adverse effects , Torsades de Pointes/chemically induced , Torsades de Pointes/diagnosis , Adult , Dose-Response Relationship, Drug , Female , Humans , Male , Potassium Channel Blockers/administration & dosage , Reproducibility of Results , Sensitivity and Specificity , Sex Factors
17.
J Mol Cell Cardiol ; 72: 126-37, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24631769

ABSTRACT

Accurate diagnosis of predisposition to long QT syndrome is crucial for reducing the risk of cardiac arrhythmias. In recent years, drug-induced provocative tests have proved useful to unmask some latent mutations linked to cardiac arrhythmias. In this study we expanded this concept by developing a prototype for a computational provocative screening test to reveal genetic predisposition to acquired long-QT syndrome (aLQTS). We developed a computational approach to reveal the pharmacological properties of IKr blocking drugs that are most likely to cause aLQTS in the setting of subtle alterations in IKr channel gating that would be expected to result from benign genetic variants. We used the model to predict the most potentially lethal combinations of kinetic anomalies and drug properties. In doing so, we also implicitly predicted ideal inverse therapeutic properties of K channel openers that would be expected to remedy a specific defect. We systematically performed "in silico mutagenesis" by altering discrete kinetic transition rates of the Fink et al. Markov model of human IKr channels, corresponding to activation, inactivation, deactivation and recovery from inactivation of IKr channels. We then screened and identified the properties of IKr blockers that caused acquired long QT and therefore unmasked mutant phenotypes for mild, moderate and severe variants. Mutant IKr channels were incorporated into the O'Hara et al. human ventricular action potential (AP) model and subjected to simulated application of a wide variety of IKr-drug interactions in order to identify the characteristics that selectively exacerbate the AP duration (APD) differences between wild-type and IKr mutated cells. Our results show that drugs with disparate affinities to conformation states of the IKr channel are key to amplify variants underlying susceptibility to acquired long QT syndrome, an effect that is especially pronounced at slow frequencies. Finally, we developed a mathematical formulation of the M54T MiRP1 latent mutation and simulated a provocative test. In this setting, application of dofetilide dramatically amplified the predicted QT interval duration in the M54T hMiRP1 mutation compared to wild-type.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Heart Ventricles/metabolism , Long QT Syndrome/metabolism , Models, Statistical , Potassium Channel Blockers/adverse effects , Potassium Channels, Voltage-Gated/agonists , Action Potentials/drug effects , Astemizole/adverse effects , Cisapride/adverse effects , Computer Simulation , Gene Expression , Genetic Predisposition to Disease , Heart Ventricles/drug effects , Heart Ventricles/pathology , Humans , Ion Channel Gating/drug effects , Kinetics , Long QT Syndrome/chemically induced , Long QT Syndrome/genetics , Long QT Syndrome/pathology , Mutation , Phenethylamines/adverse effects , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Protein Conformation , Severity of Illness Index , Sotalol/adverse effects , Sulfonamides/adverse effects , Terfenadine/adverse effects
18.
Br J Anaesth ; 113(5): 875-83, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24989775

ABSTRACT

BACKGROUND: Potassium-channels in the carotid body and the brainstem are important regulators of ventilation. The BKCa-channel contains response elements for CO, O2, and CO2. Its block increases carotid body signalling, phrenic nerve activity, and respiratory drive. GAL-021, a new BKCa-channel blocker, increases minute ventilation in rats and non-human primates. This study assessed the single-dose safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of GAL-021 in healthy volunteers. METHODS: Thirty subjects participated in a nine-period, randomized, double-blinded, placebo-controlled, crossover, ascending dose, first-in-human study with i.v. infusions of 0.1-0.96 mg kg(-1) h(-1) for 1 h and intermediate doses up to 4 h. RESULTS: Adverse event rates were generally similar among dose levels and between placebo- and GAL-021-treated subjects. At higher GAL-021 doses, a mild/moderate burning sensation at the infusion site occurred during the infusion. No clinically significant changes in vital signs or clinical chemistries were noted. Minute ventilation increased (AUE0-1 h ≈ 16%, P<0.05) and end-tidal carbon dioxide ([Formula: see text]) decreased (AUE0-1 h ≈ 6%, P<0.05) during the first hour at 0.96 mg kg(-1) h(-1) with 1/2-maximal [Formula: see text] and [Formula: see text]-change occurring by 7.5 min. Drug concentration rose rapidly during the infusion and decreased rapidly initially (distribution t1/2 of 30 min) and then more slowly (terminal t1/2 of 5.6 h). CONCLUSIONS: GAL-021 was safe and generally well tolerated with adverse events comparable with placebo except for an infusion site burning sensation. GAL-021 stimulated ventilation at the highest doses suggesting that greater infusion rates may be required for maximum PD effects. GAL-021 had PK characteristics consistent with an acute care medication.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Triazines/pharmacology , Adult , Carbon Dioxide/blood , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Infusions, Intravenous , Male , Potassium Channel Blockers/adverse effects , Potassium Channel Blockers/pharmacokinetics , Respiratory Mechanics/drug effects , Triazines/adverse effects , Triazines/pharmacokinetics , Young Adult
20.
Planta Med ; 80(12): 1045-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25089737

ABSTRACT

Inhibition of the human ether-a-go-go-related gene channel is the single most important risk factor leading to acquired long QT syndrome. Drug-induced QT prolongation can cause severe cardiac complications, including arrhythmia, and is thus a liability in drug development. Considering the importance of the human ether-a-go-go-related gene channel as an antitarget and the daily intake of plant-derived foods and herbal products, surprisingly few natural products have been tested for channel blocking properties. In an assessment of possible human ether-a-go-go-related gene liabilities, a selection of widely used herbal medicines and edible plants (vegetables, fruits, and spices) was screened by means of a functional two-microelectrode voltage-clamp assay with Xenopus oocytes. The human ether-a-go-go-related gene channel blocking activity of selected extracts was investigated with the aid of a high-performance liquid chromatography-based profiling approach, and attributed to tannins and alkaloids. Major European medicinal plants and frequently consumed food plants were found to have a low risk for human ether-a-go-go-related gene toxicity.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Heart Conduction System/abnormalities , Plant Extracts/pharmacology , Plants, Edible/chemistry , Plants, Medicinal/chemistry , Potassium Channel Blockers/pharmacology , Alkaloids/adverse effects , Alkaloids/pharmacology , Animals , Biological Products , Brugada Syndrome , Cardiac Conduction System Disease , Herbal Medicine , Humans , Oocytes , Plant Extracts/adverse effects , Potassium Channel Blockers/adverse effects , Tannins/adverse effects , Tannins/pharmacology , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL