1
|
Murai K, Vasigh A, Alexy T, Tóth K, Czopf L. The Role of Ranolazine in the Treatment of Ventricular Tachycardia and Atrial Fibrillation: A Narrative Review of the Clinical Evidence. Biomedicines 2024; 12:1669. [PMID: 39200134 PMCID: PMC11351540 DOI: 10.3390/biomedicines12081669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Cardiac arrhythmias are among the leading causes of morbidity and mortality worldwide. While antiarrhythmic drugs traditionally represent the first-line management strategy, their use is often limited by profound proarrhythmic effects. Several studies, including randomized control trials (RCTs), have demonstrated the antiarrhythmic efficacy of ranolazine, which is registered as an antianginal agent, while also establishing its safety profile. This review compiles clinical evidence investigating the antiarrhythmic properties of ranolazine, focusing primarily on ventricular tachycardia (VT) and atrial fibrillation (AF), as they are common rhythm abnormalities with serious complications. Data from RCTs indicate that ranolazine reduces VT incidence, although this effect is not universal. Therefore, we attempt to better describe the patient population that gains the most benefit from ranolazine due to VT suppression. Additionally, ranolazine is known to enhance the conversion rate of AF to sinus rhythm when combined with other antiarrhythmic drugs such as amiodarone, highlighting its synergistic effect in the atrium without provoking ventricular dysrhythmias. Despite the heterogeneity in the currently available data, ranolazine appears to be an effective and safe option for the management of various arrhythmias.
Collapse
Affiliation(s)
- Kyosuke Murai
- 1st Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary (K.T.)
| | - Amir Vasigh
- 1st Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary (K.T.)
| | - Tamás Alexy
- Department of Medicine, Division of Cardiology, University of Minnesota, Minneapolis, MN 55127, USA;
| | - Kálmán Tóth
- 1st Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary (K.T.)
- Szentágothai János Research Centre, Medical School, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - László Czopf
- 1st Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary (K.T.)
| |
Collapse
|
2
|
Song T, Hui W, Huang M, Guo Y, Yu M, Yang X, Liu Y, Chen X. Dynamic Changes in Ion Channels during Myocardial Infarction and Therapeutic Challenges. Int J Mol Sci 2024; 25:6467. [PMID: 38928173 PMCID: PMC11203447 DOI: 10.3390/ijms25126467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
In different areas of the heart, action potential waveforms differ due to differences in the expressions of sodium, calcium, and potassium channels. One of the characteristics of myocardial infarction (MI) is an imbalance in oxygen supply and demand, leading to ion imbalance. After MI, the regulation and expression levels of K+, Ca2+, and Na+ ion channels in cardiomyocytes are altered, which affects the regularity of cardiac rhythm and leads to myocardial injury. Myocardial fibroblasts are the main effector cells in the process of MI repair. The ion channels of myocardial fibroblasts play an important role in the process of MI. At the same time, a large number of ion channels are expressed in immune cells, which play an important role by regulating the in- and outflow of ions to complete intracellular signal transduction. Ion channels are widely distributed in a variety of cells and are attractive targets for drug development. This article reviews the changes in different ion channels after MI and the therapeutic drugs for these channels. We analyze the complex molecular mechanisms behind myocardial ion channel regulation and the challenges in ion channel drug therapy.
Collapse
Affiliation(s)
- Tongtong Song
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
- Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130012, China
| | - Wenting Hui
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Min Huang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Yan Guo
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Meiyi Yu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Xiaoyu Yang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Yanqing Liu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China; (T.S.); (W.H.); (M.H.); (Y.G.); (M.Y.); (X.Y.); (Y.L.)
| |
Collapse
|
3
|
Djamgoz MBA. Ranolazine: a potential anti-metastatic drug targeting voltage-gated sodium channels. Br J Cancer 2024; 130:1415-1419. [PMID: 38424164 PMCID: PMC11058819 DOI: 10.1038/s41416-024-02622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Multi-faceted evidence from a range of cancers suggests strongly that de novo expression of voltage-gated sodium channels (VGSCs) plays a significant role in driving cancer cell invasiveness. Under hypoxic conditions, common to growing tumours, VGSCs develop a persistent current (INaP) which can be blocked selectively by ranolazine. METHODS Several different carcinomas were examined. We used data from a range of experimental approaches relating to cellular invasiveness and metastasis. These were supplemented by survival data mined from cancer patients. RESULTS In vitro, ranolazine inhibited invasiveness of cancer cells especially under hypoxia. In vivo, ranolazine suppressed the metastatic abilities of breast and prostate cancers and melanoma. These data were supported by a major retrospective epidemiological study on breast, colon and prostate cancer patients. This showed that risk of dying from cancer was reduced by ca.60% among those taking ranolazine, even if this started 4 years after the diagnosis. Ranolazine was also shown to reduce the adverse effects of chemotherapy on heart and brain. Furthermore, its anti-cancer effectiveness could be boosted by co-administration with other drugs. CONCLUSIONS Ranolazine, alone or in combination with appropriate therapies, could be reformulated as a safe anti-metastatic drug offering many potential advantages over current systemic treatment modalities.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
- Biotechnology Research Centre, Cyprus International University, Haspolat, Nicosia, TRNC, Mersin, 10, Türkiye.
| |
Collapse
|
4
|
Delayed Ventricular Repolarization and Sodium Channel Current Modification in a Mouse Model of Rett Syndrome. Int J Mol Sci 2022; 23:ijms23105735. [PMID: 35628543 PMCID: PMC9147596 DOI: 10.3390/ijms23105735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Rett syndrome (RTT) is a severe developmental disorder that is strongly linked to mutations in the MECP2 gene. RTT has been associated with sudden unexplained death and ECG QT interval prolongation. There are mixed reports regarding QT prolongation in mouse models of RTT, with some evidence that loss of Mecp2 function enhances cardiac late Na current, INa,Late. The present study was undertaken in order to investigate both ECG and ventricular AP characteristics in the Mecp2Null/Y male murine RTT model and to interrogate both fast INa and INa,Late in myocytes from the model. ECG recordings from 8-10-week-old Mecp2Null/Y male mice revealed prolongation of the QT and rate corrected QT (QTc) intervals and QRS widening compared to wild-type (WT) controls. Action potentials (APs) from Mecp2Null/Y myocytes exhibited longer APD75 and APD90 values, increased triangulation and instability. INa,Late was also significantly larger in Mecp2Null/Y than WT myocytes and was insensitive to the Nav1.8 inhibitor A-803467. Selective recordings of fast INa revealed a decrease in peak current amplitude without significant voltage shifts in activation or inactivation V0.5. Fast INa 'window current' was reduced in RTT myocytes; small but significant alterations of inactivation and reactivation time-courses were detected. Effects of two INa,Late inhibitors, ranolazine and GS-6615 (eleclazine), were investigated. Treatment with 30 µM ranolazine produced similar levels of inhibition of INa,Late in WT and Mecp2Null/Y myocytes, but produced ventricular AP prolongation not abbreviation. In contrast, 10 µM GS-6615 both inhibited INa,Late and shortened ventricular AP duration. The observed changes in INa and INa,Late can account for the corresponding ECG changes in this RTT model. GS-6615 merits further investigation as a potential treatment for QT prolongation in RTT.
Collapse
|
5
|
Antiarrhythmic Agents: a Review and Comment on Relevance in the Current Era—Part 2. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2021. [DOI: 10.1007/s11936-021-00944-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
6
|
Nabeh OA, Helaly MM, Menshawey R, Menshawey E, Nasser MMM, Diaa El-deen AM. Contemporary approach to understand and manage COVID-19-related arrhythmia. Egypt Heart J 2021; 73:76. [PMID: 34459992 PMCID: PMC8403826 DOI: 10.1186/s43044-021-00201-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
Arrhythmia, one of the most common complications of COVID-19, was reported in nearly one-third of diagnosed COVID-19 patients, with higher prevalence rate among ICU admitted patients. The underlying etiology for arrhythmia in these cases are mostly multifactorial as those patients may suffer from one or more of the following predisposing mechanisms; catecholamine surge, hypoxia, myocarditis, cytokine storm, QTc prolongation, electrolyte disturbance, and pro-arrhythmic drugs usage. Obviously, the risk for arrhythmia and the associated lethal outcome would rise dramatically among patients with preexisting cardiac disease such as myocardial ischemia, heart failure, cardiomyopathy, and hereditary arrhythmias. Considering all of these variables, the management strategy of COVID-19 patients should expand from managing a viral infection and related host immune response to include the prevention of predictable causes for arrhythmia. This may necessitate the need to investigate the role of some drugs that modulate the pathway of arrhythmia generation. Of these drugs, we discuss the potential role of adrenergic antagonists, trimetazidine, ranolazine, and the debatable angiotensin converting enzyme inhibitors drugs. We also recommend monitoring the level of: unbound free fatty acids, serum electrolytes, troponin, and QTc (even in the absence of apparent pro-arrhythmic drug use) as these may be the only indicators for patients at risk for arrhythmic complications.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maiada Mohamed Helaly
- Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rahma Menshawey
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Esraa Menshawey
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
7
|
Huang M, Liao Z, Li X, Yang Z, Fan X, Li Y, Zhao Z, Lang S, Cyganek L, Zhou X, Akin I, Borggrefe M, El-Battrawy I. Effects of Antiarrhythmic Drugs on hERG Gating in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes From a Patient With Short QT Syndrome Type 1. Front Pharmacol 2021; 12:675003. [PMID: 34025432 PMCID: PMC8138577 DOI: 10.3389/fphar.2021.675003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Aims: The short QT syndrome type 1 (SQT1) is linked to hERG channel mutations (e.g., N588K). Drug effects on hERG channel gating kinetics in SQT1-cells have not been investigated. Methods: This study used hiPSC-CMs of a healthy donor and a SQT1-patient carrying the N588K mutation and patch clamp to examine the drug effects on hERG channel gating kinetics. Results: Ajmaline, amiodarone, ivabradine, flecainide, quinidine, mexiletine and ranolazine inhibited the hERG channel current (IKr) less strongly in hiPSC-CMs from the SQTS1-patient (SQT1-hiPSC-CMs) comparing with cells from the healthy donor (donor-hiPSC-CMs). Quinidine and mexiletine reduced, but ajmaline, amiodarone, ivabradine and ranolazine increased the time to peak of IKr similarly in SQT1-hiPSC-CMs and donor-hiPSC-CMs. Although regarding the shift of activation and inactivation curves, tested drugs showed differential effects in donor- and SQT1-hiPSC-CMs, quinidine, ajmaline, ivabradine and mexiletine but not amiodarone, flecainide and ranolazine reduced the window current in SQT1-hiPSC-CMs. Quinidine, ajmaline, ivabradine and mexiletine differentially changed the time constant of recovery from inactivation, but all of them increased the time constant of deactivation in SQT1-hiPSC-CMs. Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. This information may be helpful for selecting drugs for treating SQT1-patients with hERG channel mutation.
Collapse
Affiliation(s)
- Mengying Huang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhenxing Liao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xin Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Yang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xuehui Fan
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yingrui Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhihan Zhao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| |
Collapse
|
8
|
Electrophysiological effects of ranolazine in a goat model of lone atrial fibrillation. Heart Rhythm 2020; 18:615-622. [PMID: 33232809 DOI: 10.1016/j.hrthm.2020.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND There is still an unmet need for pharmacologic treatment of atrial fibrillation (AF) with few effects on ventricular electrophysiology. Ranolazine is an antiarrhythmic drug reported to have strong atrial selectivity. OBJECTIVE The purpose of this study was to investigate the electrophysiological effects of ranolazine in atria with AF-induced electrical remodeling in a model of lone AF in awake goats. METHODS Electrode patches were implanted on the atrial epicardium of 8 Dutch milk goats. Experiments were performed at baseline and after 2 and 14 days of electrically maintained AF. Several electrophysiological parameters and AF episode duration were measured during infusion of vehicle and different doses of ranolazine (target plasma levels 4, 8, and 16 μM, respectively). RESULTS The highest dose of ranolazine significantly prolonged atrial effective refractory period and decreased atrial conduction velocity at baseline and after 2 days of AF. After 2 weeks of AF, ranolazine prolonged the p5 and p50 of AF cycle length distribution in a dose-dependent manner but was not effective in restoring sinus rhythm. No adverse ventricular arrhythmic events (eg, premature ventricular beats or signs of hemodynamic instability) were observed during infusion of ranolazine at any point in the study. CONCLUSION The lowest investigated dose of ranolazine, which is expected to block both late INa and atrial peak INa, had no effect on the investigated electrophysiological parameters. The highest dose affected both atrial and ventricular electrophysiological parameters at different stages of AF-induced remodeling but was not efficacious in cardioverting AF to sinus rhythm in a goat model of lone AF.
Collapse
|
9
|
Bentzen BH, Bomholtz SH, Simó-Vicens R, Folkersen L, Abildgaard L, Speerschneider T, Muthukumarasamy KM, Edvardsson N, Sørensen US, Grunnet M, Diness JG. Mechanisms of Action of the KCa2-Negative Modulator AP30663, a Novel Compound in Development for Treatment of Atrial Fibrillation in Man. Front Pharmacol 2020; 11:610. [PMID: 32477117 PMCID: PMC7232560 DOI: 10.3389/fphar.2020.00610] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/20/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Small conductance Ca2+-activated K+ channels (SK channels, KCa2) are a new target for treatment of atrial fibrillation (AF). AP30663 is a small molecule inhibitor of KCa2 channels that is currently in clinical development for treatment of AF. The aim of this study is to present the electrophysiological profile and mechanism of action of AP30663 and its efficacy in prolonging atrial refractoriness in rodents, and by bioinformatic analysis investigate if genetic variants in KCNN2 or KCNN3 influence the expression level of these in human heart tissue. METHODS AND RESULTS Whole-cell and inside-out patch-clamp recordings of heterologously expressed KCa2 channels revealed that AP30663 inhibits KCa2 channels with minor effects on other relevant cardiac ion channels. AP30663 modulates the KCa2.3 channel by right-shifting the Ca2+-activation curve. In isolated guinea pig hearts AP30663 significantly prolonged the atrial effective refractory period (AERP) with minor effects on the QT-interval corrected for heart rate. Similarly, in anaesthetized rats 5 and 10 mg/kg of AP30663 changed the AERP to 130.7±5.4% and 189.9±18.6 of baseline values. The expression quantitative trait loci analyses revealed that the genome wide association studies for AF SNP rs13376333 in KCNN3 is associated with increased mRNA expression of KCNN3 in human atrial appendage tissue. CONCLUSIONS AP30663 is a novel negative allosteric modulator of KCa2 channels that concentration-dependently prolonged rodent atrial refractoriness with minor effects on the QT-interval. Moreover, AF associated SNPs in KCNN3 influence KCNN3 mRNA expression in human atrial tissue. These properties support continued development of AP30663 for treatment of AF in man.
Collapse
Affiliation(s)
- Bo Hjorth Bentzen
- Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofia Hammami Bomholtz
- Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rafel Simó-Vicens
- Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Folkersen
- Institute of Biological Psychiatry, Sankt Hans Hospital, Roskilde, Denmark
| | | | - Tobias Speerschneider
- Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kalai Mangai Muthukumarasamy
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nils Edvardsson
- Acesion Pharma, Copenhagen, Denmark
- Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | |
Collapse
|
10
|
Takla M, Huang CLH, Jeevaratnam K. The cardiac CaMKII-Na v1.5 relationship: From physiology to pathology. J Mol Cell Cardiol 2020; 139:190-200. [PMID: 31958466 DOI: 10.1016/j.yjmcc.2019.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/20/2019] [Accepted: 12/30/2019] [Indexed: 12/19/2022]
Abstract
The SCN5A gene encodes Nav1.5, which, as the cardiac voltage-gated Na+ channel's pore-forming α subunit, is crucial for the initiation and propagation of atrial and ventricular action potentials. The arrhythmogenic propensity of inherited SCN5A mutations implicates the Na+ channel in determining cardiomyocyte excitability under normal conditions. Cytosolic kinases have long been known to alter the kinetic profile of Nav1.5 inactivation via phosphorylation of specific residues. Recent substantiation of both the role of calmodulin-dependent kinase II (CaMKII) in modulating the properties of the Nav1.5 inactivation gate and the significant rise in oxidation-dependent autonomous CaMKII activity in structural heart disease has raised the possibility of a novel pathway for acquired arrhythmias - the CaMKII-Nav1.5 relationship. The aim of this review is to: (1) outline the relationship's translation from physiological adaptation to pathological vicious circle; and (2) discuss the relative merits of each of its components as pharmacological targets.
Collapse
Affiliation(s)
- Michael Takla
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom
| | - Christopher L-H Huang
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom.
| |
Collapse
|
11
|
Carstensen H, Hesselkilde EZ, Haugaard MM, Flethøj M, Carlson J, Pehrson S, Jespersen T, Platonov PG, Buhl R. Effects of dofetilide and ranolazine on atrial fibrillatory rate in a horse model of acutely induced atrial fibrillation. J Cardiovasc Electrophysiol 2019; 30:596-606. [PMID: 30661267 PMCID: PMC6849868 DOI: 10.1111/jce.13849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The atrial fibrillatory rate is a potential biomarker in the study of antiarrhythmic drug effects on atrial fibrillation (AF). The purpose of this study was to evaluate whether dose-dependent changes in the atrial fibrillatory rate can be monitored on surface electrocardiography (ECG) following treatment with dofetilide, ranolazine, and a combination of the two in an acute model of AF in horses. METHODS AND RESULTS Eight horses were subjected to pacing-induced AF on 4 separate days. Saline (control), dofetilide, ranolazine, or a combination of dofetilide and ranolazine was administered in four incremental doses. Atrial fibrillatory activity was extracted from surface ECGs using spatiotemporal QRST cancellation. The mean atrial fibrillatory rate before drug infusion was 297 ± 27 fpm. Dofetilide reduced the atrial fibrillatory rate following the infusion of low doses (0.89 µg/kg, P < 0.05) and within 5 minutes preceding cardioversion (P < 0.05). Cardioversion with ranolazine was preceded by a reduction in the atrial fibrillatory rate in the last minute (P < 0.05). The combination of drugs reduced the atrial fibrillatory rate in a similar manner to dofetilide used alone. A trend toward a lower atrial fibrillatory rate before drug infusion was found among horses cardioverting on low doses of the drugs. CONCLUSION The atrial fibrillatory rate derived from surface ECGs showed a difference in the mode of action on AF between dofetilide and ranolazine. Dofetilide reduced the atrial fibrillatory rate, whereas ranolazine displayed a cardioverting mechanism that was distinct from a slowing of the fibrillatory process.
Collapse
Affiliation(s)
- Helena Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Eva Zander Hesselkilde
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Maria Mathilde Haugaard
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Mette Flethøj
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Jonas Carlson
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Steen Pehrson
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Taastrup, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Pyotr G Platonov
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.,Arrhythmia Clinic, Skåne University Hospital, Lund, Sweden
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
12
|
Antiarrhythmic Effects of Combining Dofetilide and Ranolazine in a Model of Acutely Induced Atrial Fibrillation in Horses. J Cardiovasc Pharmacol 2019; 71:26-35. [PMID: 29068807 PMCID: PMC5768216 DOI: 10.1097/fjc.0000000000000541] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is Available in the Text. Background: Antiarrhythmic compounds against atrial fibrillation (AF) often have reduced efficacy and may display cardiac and/or noncardiac toxicity. Efficacy can be improved by combining 2 compounds with distinct mechanisms, and it may be possible to use lower doses of each compound, thereby reducing the likelihood of adverse side effects. The purpose of this study was to investigate whether the effective doses of dofetilide and ranolazine can be reduced if the drugs are combined. Methods: Dofetilide, ranolazine, and a combination of these were administered in 4 incremental dosing regimens to horses with acutely pacing-induced AF. Time to cardioversion, atrial effective refractory period, and AF vulnerability and duration were assessed. Results: Of 8 horses, 6 cardioverted to sinus rhythm after infusion with a combination of 0.889 μg/kg dofetilide and 0.104 mg/kg ranolazine. Two horses cardioverted with 0.104 mg/kg ranolazine alone, and 3 cardioverted with 0.889 μg/kg dofetilide alone. The combination therapy decreased AF vulnerability (P < 0.05) and AF duration (P < 0.05). No change in atrial effective refractory period was detected with any of the drugs. Conclusions: The combination of dofetilide and ranolazine showed increased antiarrhythmic effects on acutely induced AF in horses, affecting time to cardioversion, AF vulnerability, and AF duration.
Collapse
|
13
|
Mezincescu A, Karthikeyan VJ, Nadar SK. Ranolazine: A true pluripotent cardiovascular drug or jack of all trades, master of none? Sultan Qaboos Univ Med J 2018; 18:e13-e23. [PMID: 29666676 DOI: 10.18295/squmj.2018.18.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/18/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Although the majority of patients with CVD are treated with interventional procedures, a substantial number require medical therapy in terms of both prognosis and symptomatic relief. However, commonly used agents such as β-blockers and calcium channel blockers reduce blood pressure in patients whose resting pressures are often already low. Ranolazine is a promising agent that does not have significant effects on blood pressure or heart rate. Use of this drug has been documented in various cardiovascular conditions, including ischaemic heart disease, heart failure and arrhythmias. This review article aimed to examine current evidence on the use of ranolazine in various cardiovascular conditions in order to determine whether it is a true pluripotent cardiovascular agent or, on the other hand, a "jack of all trades, master of none."
Collapse
Affiliation(s)
- Alice Mezincescu
- Cardiovascular & Diabetes Research Unit, University of Aberdeen, Aberdeen, UK
| | - V J Karthikeyan
- Royal Albert Edward Infirmary, Wrightington Wigan & Leigh NHS Foundation Trust, Wigan, UK
| | - Sunil K Nadar
- Department of Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
14
|
Polak S, Romero K, Berg A, Patel N, Jamei M, Hermann D, Hanna D. Quantitative approach for cardiac risk assessment and interpretation in tuberculosis drug development. J Pharmacokinet Pharmacodyn 2018. [PMID: 29520534 PMCID: PMC5953981 DOI: 10.1007/s10928-018-9580-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
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.
Collapse
Affiliation(s)
- Sebastian Polak
- Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.,Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków, 30-217, Poland
| | - Klaus Romero
- Critical Path Institute, 1730 E. River Road, Tucson, AZ, 85705, USA.
| | - Alexander Berg
- Critical Path Institute, 1730 E. River Road, Tucson, AZ, 85705, USA
| | - Nikunjkumar Patel
- Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Masoud Jamei
- Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | | | - Debra Hanna
- Critical Path Institute, 1730 E. River Road, Tucson, AZ, 85705, USA
| |
Collapse
|
15
|
Abdelsayed M, Ruprai M, Ruben PC. The efficacy of Ranolazine on E1784K is altered by temperature and calcium. Sci Rep 2018; 8:3643. [PMID: 29483621 PMCID: PMC5827758 DOI: 10.1038/s41598-018-22033-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/15/2018] [Indexed: 12/19/2022] Open
Abstract
E1784K is the most common mixed syndrome SCN5a mutation underpinning both Brugada syndrome type 1 (BrS1) and Long-QT syndrome type 3 (LQT3). The charge reversal mutant enhances the late sodium current (INa) passed by the cardiac voltage-gated sodium channel (NaV1.5), delaying cardiac repolarization. Exercise-induced triggers, like elevated temperature and cytosolic calcium, exacerbate E1784K late INa. In this study, we tested the effects of Ranolazine, the late INa blocker, on voltage-dependent and kinetic properties of E1784K at elevated temperature and cytosolic calcium. We used whole-cell patch clamp to measure INa from wild type and E1784K channels expressed in HEK293 cells. At elevated temperature, Ranolazine attenuated gain-of-function in E1784K by decreasing late INa, hyperpolarizing steady-state fast inactivation, and increasing use-dependent inactivation. Both elevated temperature and cytosolic calcium hampered the capacity of Ranolazine to suppress E1784K late INa. In-silico action potential (AP) simulations were done using a modified O'Hara Rudy (ORd) cardiac model. Simulations showed that Ranolazine failed to shorten AP duration, an effect augmented at febrile temperatures. The drug-channel interaction is clearly affected by external triggers, as reported previously with ischemia. Determining drug efficacy under various physiological states in SCN5a cohorts is crucial for accurate management of arrhythmias.
Collapse
Affiliation(s)
- Mena Abdelsayed
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Manpreet Ruprai
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Cardiomyopathies due to genetic mutations are a heterogeneous group of disorders that comprise diseases of contractility, myocardial relaxation, and arrhythmias. Our goal here is to discuss a limited list of genetically inherited cardiomyopathies and the specific therapeutic strategies used to treat them. RECENT FINDINGS Research into the molecular pathophysiology of the development of these cardiomyopathies is leading to the development of novel treatment approaches. Therapies targeting these specific mutations with gene therapy vectors are on the horizon, while other therapies which indirectly affect the physiologic derangements of the mutations are currently being studied and used clinically. Many of these therapies are older medications being given new roles such as mexiletine for Brugada syndrome and diflunisal for transthyretin amyloid cardiomyopathy. A newer targeted therapy, the inhibitor of myosin ATPase MYK-461, has been shown to suppress the development of ventricular hypertrophy, fibrosis, and myocyte disarray and is being studied as a potential therapy in patients with hypertrophic cardiomyopathy. While this field is too large to be completely contained in a single review, we present a large cross section of recent developments in the field of therapeutics for inherited cardiomyopathies. New therapies are on the horizon, and their development will likely result in improved outcomes for patients inflicted by these conditions.
Collapse
Affiliation(s)
- Kenneth Varian
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA. .,Center for Clinical Genomics, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
17
|
Late sodium current associated cardiac electrophysiological and mechanical dysfunction. Pflugers Arch 2017; 470:461-469. [DOI: 10.1007/s00424-017-2079-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
18
|
|
19
|
Azam MA, Zamiri N, Massé S, Kusha M, Lai PFH, Nair GK, Tan NS, Labos C, Nanthakumar K. Effects of Late Sodium Current Blockade on Ventricular Refibrillation in a Rabbit Model. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.116.004331. [PMID: 28314848 DOI: 10.1161/circep.116.004331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND After defibrillation of initial ventricular fibrillation (VF), it is crucial to prevent refibrillation to ensure successful resuscitation outcomes. Inability of the late Na+ current to inactivate leads to intracellular Ca2+ dysregulation and arrhythmias. Our aim was to determine the effects of ranolazine and GS-967, inhibitors of the late Na+ current, on ventricular refibrillation. METHODS AND RESULTS Long-duration VF was induced electrically in Langendorff-perfused rabbit hearts (n=22) and terminated with a defibrillator after 6 minutes. Fibrillating hearts were randomized into 3 groups: treatment with ranolazine, GS-967, or nontreated controls. In the treated groups, hearts were perfused with ranolazine or GS-967 at 2 minutes of VF. In control experiments, perfusion solution was supplemented with isotonic saline in lieu of a drug. Inducibility of refibrillation was assessed after initial long-duration VF by attempting to reinduce VF. Sustained refibrillation was successful in fewer ranolazine-treated (29.17%; P=0.005) or GS-967-treated (45.83%, P=0.035) hearts compared with that in nontreated control hearts (84.85%). In GS-967-treated hearts, significantly more spontaneous termination of initial long-duration VF was observed (66.67%; P=0.01). Ca2+ transient duration was reduced in ranolazine-treated hearts compared with that in controls (P=0.05) and also Ca2+ alternans (P=0.03). CONCLUSIONS Late Na+ current inhibition during long-duration VF reduces the susceptibility to subsequent refibrillation, partially by mitigating dysregulation of intracellular Ca2+. These results suggest the potential therapeutic use of ranolazine and GS-967 and call for further testing in cardiac arrest models.
Collapse
Affiliation(s)
- Mohammed Ali Azam
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nima Zamiri
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Stéphane Massé
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Marjan Kusha
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Patrick F H Lai
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Govind K Nair
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nigel S Tan
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Christopher Labos
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Kumaraswamy Nanthakumar
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.).
| |
Collapse
|
20
|
Chorin E, Hu D, Antzelevitch C, Hochstadt A, Belardinelli L, Zeltser D, Barajas-Martinez H, Rozovski U, Rosso R, Adler A, Benhorin J, Viskin S. Ranolazine for Congenital Long-QT Syndrome Type III: Experimental and Long-Term Clinical Data. Circ Arrhythm Electrophysiol 2017; 9:CIRCEP.116.004370. [PMID: 27733495 DOI: 10.1161/circep.116.004370] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/29/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND The basic defect in long-QT syndrome type III (LQT3) is an excessive inflow of sodium current during phase 3 of the action potential caused by mutations in the SCN5A gene. Most sodium channel blockers reduce the early (peak) and late components of the sodium current (INa and INaL), but ranolazine preferentially reduces INaL. We, therefore, evaluated the effects of ranolazine in LQT3 caused by the D1790G mutation in SCN5A. METHODS AND RESULTS We performed an experimental study of ranolazine in TSA201 cells expressing the D1790G mutation. We then performed a long-term clinical evaluation of ranolazine in LQT3 patients carrying the D1790G mutation. In the experimental study, INaL was significantly higher in D1790G than in wild-type channels expressed in the TSA201 cells. Ranolazine exerted a concentration-dependent block of INaL of the SCN5A-D1790G channel without reducing peak INa significantly. In the clinical study, among 8 patients with LQT3 and confirmed D1790G mutation, ranolazine had no effects on the sinus rate or QRS width but shortened the QTc from 509±41 to 451±26 ms, a mean decrease of 56±52 ms (10.6%; P=0.012). The QT-shortening effect of ranolazine remained effective throughout the entire study period of 22.8±12.8 months. Ranolazine reduced the QTc at all heart rates but less so during extreme nocturnal bradycardia. A type I Brugada ECG was never noticed. CONCLUSIONS Ranolazine blocks INaL in experimental models of LQT3 harboring the SCN5A-D1790G mutation and shortened the QT interval of LQT3 patients. CLINICAL TRIAL REGISTRATION URL: https://clinicaltrials.gov; Unique identifier: NCT01728025.
Collapse
Affiliation(s)
- Ehud Chorin
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Dan Hu
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Charles Antzelevitch
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Aviram Hochstadt
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Luiz Belardinelli
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - David Zeltser
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Hector Barajas-Martinez
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Uri Rozovski
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Raphael Rosso
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Arnon Adler
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Jesaia Benhorin
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.)
| | - Sami Viskin
- From the Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Israel (E.C., A.H., D.Z., U.R., R.R., A.A., J.B., S.V.); Masonic Medical Research Laboratory, Utica, NY (D.H., C.A., H.B.-M.); Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, PA (C.A.); and Gilead Sciences, Inc, Foster City, CA (L.B.).
| |
Collapse
|
21
|
Wiśniowska B, Polak S. Am I or am I not proarrhythmic? Comparison of various classifications of drug TdP propensity. Drug Discov Today 2017; 22:10-16. [DOI: 10.1016/j.drudis.2016.09.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/22/2016] [Accepted: 09/28/2016] [Indexed: 12/12/2022]
|
22
|
Tanimura A, Lim SAO, Aceves Buendia JDJ, Goldberg JA, Surmeier DJ. Cholinergic Interneurons Amplify Corticostriatal Synaptic Responses in the Q175 Model of Huntington's Disease. Front Syst Neurosci 2016; 10:102. [PMID: 28018188 PMCID: PMC5159611 DOI: 10.3389/fnsys.2016.00102] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/02/2016] [Indexed: 01/18/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by deficits in movement control that are widely viewed as stemming from pathophysiological changes in the striatum. Giant, aspiny cholinergic interneurons (ChIs) are key elements in the striatal circuitry controlling movement, but whether their physiological properties are intact in the HD brain is unclear. To address this issue, the synaptic properties of ChIs were examined using optogenetic approaches in the Q175 mouse model of HD. In ex vivo brain slices, synaptic facilitation at thalamostriatal synapses onto ChIs was reduced in Q175 mice. The alteration in thalamostriatal transmission was paralleled by an increased response to optogenetic stimulation of cortical axons, enabling these inputs to more readily induce burst-pause patterns of activity in ChIs. This adaptation was dependent upon amplification of cortically evoked responses by a post-synaptic upregulation of voltage-dependent Na+ channels. This upregulation also led to an increased ability of somatic spikes to invade ChI dendrites. However, there was not an alteration in the basal pacemaking rate of ChIs, possibly due to increased availability of Kv4 channels. Thus, there is a functional "re-wiring" of the striatal networks in Q175 mice, which results in greater cortical control of phasic ChI activity, which is widely thought to shape the impact of salient stimuli on striatal action selection.
Collapse
Affiliation(s)
- Asami Tanimura
- Department of Physiology, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| | - Sean Austin O Lim
- Department of Physiology, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| | - Jose de Jesus Aceves Buendia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem, Israel
| | - Joshua A Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem, Israel
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| |
Collapse
|
23
|
Chaitman BR. Efficacy and Safety of a Metabolic Modulator Drug in Chronic Stable Angina: Review of Evidence from Clinical Trials. J Cardiovasc Pharmacol Ther 2016; 9 Suppl 1:S47-64. [PMID: 15378131 DOI: 10.1177/107424840400900105] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A number of newer antianginal agents, including nicorandil, trimetazidine, and ivabradine, have been synthesized in recent years, but ranolazine, a piperazine derivative that partially inhibits fatty acid oxidation and the late INa current in animal models, is of particular interest mechanistically. Earlier clinical trials with immediate-release ranolazine led to the current sustained-release version tested in the Monotherapy Assessment of Ranolazine In Stable Angina (MARISA) (n = 193) and Combination Assessment of Ranolazine In Stable Angina (CARISA) trials (n = 823) of patients with chronic angina and severe limitation of exercise capacity (ie, < 5 metabolic equivalents). MARISA was a placebo-controlled, randomized trial that compared ranolazine monotherapy (500 mg, 1000 mg, and 1500 mg, twice daily) to placebo. CARISA was a placebo-controlled trial that randomized patients on background 1-blocker or calcium antagonist therapy to placebo or ranolazine (750 mg or 1000 mg, twice daily). Both studies showed a significant increase in total exercise duration, time to angina onset, and time to 1 mm ST segment depression. The average magnitude of increase in exercise duration over placebo was 29 to 56 seconds at peak and 24 to 46 seconds at trough with the 3 doses tested in MARISA, and 24 to 34 seconds greater than placebo with the 2 doses used in CARISA. The beneficial effect was achieved without clinically important changes in rest or exercise heart rate or blood pressure. Weekly angina attack frequency and nitroglycerin usage were significantly reduced in a dose-dependent manner in the 12-week CARISA trial. Reported adverse effects were similar in MARISA and CARISA and consisted of asthenia, nausea, constipation, and dizziness. Syncope, reported in 8 patients at doses of 1000 mg twice daily or more may be related to attenuation of α-1 receptor activity. The mean QTc interval increased with dose and was less than 10 msec on ranolazine at 1000 mg twice daily. The mortality rates at 1 and 2 years in MARISA and CARISA open-label run-on studies were 2% and less than 5%, acceptable for this high-risk population with limited exercise capacity. In conclusion, clinical trial evidence with ranolazine to date is consistent with its proposed mechanism of action and demonstrates an effective antianginal profile that may benefit patients with severe chronic angina.
Collapse
|
24
|
Pre- and Delayed Treatments With Ranolazine Ameliorate Ventricular Arrhythmias and Nav1.5 Downregulation in Ischemic/Reperfused Rat Hearts. J Cardiovasc Pharmacol 2016; 68:269-279. [DOI: 10.1097/fjc.0000000000000412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Aguilar M, Nattel S. The Past, Present, and Potential Future of Sodium Channel Block as an Atrial Fibrillation Suppressing Strategy. J Cardiovasc Pharmacol 2016; 66:432-40. [PMID: 25923324 DOI: 10.1097/fjc.0000000000000271] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite major advances in arrhythmia therapy, atrial fibrillation (AF) remains a challenge. A significant limitation in AF management is the lack of safe and effective drugs to restore and/or maintain sinus rhythm. The rational design of a new generation of AF-selective Na(+) channel blockers (NCBs) is emerging as a promising AF-suppressing strategy. Recent theoretical and experimental advances have generated insights into the mechanisms underlying AF maintenance and termination by antiarrhythmic drugs. Our understanding of antiarrhythmic drug-induced proarrhythmia has also grown in sophistication. These discoveries have created new possibilities in therapeutic targeting and renewed interest in improved NCB antiarrhythmic drugs. Recently described differences in atrial versus ventricular electrophysiology can be exploited in the prospective design of atrial-selective NCBs. Furthermore, state-dependent block has been shown to be an important modulator of NCB rate selectivity. Together, differential atrial-ventricular electrophysiological actions and state-dependent block form the backbone for the rational design of an AF-selective NCB. Synergistic combinations incorporating both NCB and block of K(+) currents may allow for further enhancement of AF selectivity. Future work on translating these basic research advances into the development of an optimized AF-selective NCB has the potential to provide safer and more effective pharmacotherapeutic options for AF, thereby fulfilling a major unmet clinical need.
Collapse
Affiliation(s)
- Martin Aguilar
- *Research Center, Montreal Heart Institute, Montreal, Québec, Canada; †Department of Physiology, Université de Montréal, Montreal, Québec, Canada; ‡Department of Medicine, McGill University, Montreal, Québec, Canada; §Department of Medicine, Université de Montréal, Montreal, Québec, Canada; and ¶Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada
| | | |
Collapse
|
26
|
Hancox JC, James AF, Marrion NV, Zhang H, Thomas D. Novel ion channel targets in atrial fibrillation. Expert Opin Ther Targets 2016; 20:947-58. [DOI: 10.1517/14728222.2016.1159300] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, University Walk, Bristol, UK
| | - Andrew F. James
- School of Physiology, Pharmacology and Neuroscience, University Walk, Bristol, UK
| | - Neil V. Marrion
- School of Physiology, Pharmacology and Neuroscience, University Walk, Bristol, UK
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, University of Manchester, Manchester, United Kingdom
| | - Dierk Thomas
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
27
|
Frommeyer G, Eckardt L. Drug-induced proarrhythmia: risk factors and electrophysiological mechanisms. Nat Rev Cardiol 2015; 13:36-47. [PMID: 26194552 DOI: 10.1038/nrcardio.2015.110] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug-induced ventricular tachyarrhythmias can be caused by cardiovascular drugs, noncardiovascular drugs, and even nonprescription agents. They can result in arrhythmic emergencies and sudden cardiac death. If a new arrhythmia or aggravation of an existing arrhythmia develops during therapy with a drug at a concentration usually considered not to be toxic, the situation can be defined as proarrhythmia. Various cardiovascular and noncardiovascular drugs can increase the occurrence of polymorphic ventricular tachycardia of the 'torsade de pointes' type. Antiarrhythmic drugs, antimicrobial agents, and antipsychotic and antidepressant drugs are the most important groups. Age, female sex, and structural heart disease are important risk factors for the occurrence of torsade de pointes. Genetic predisposition and individual pharmacodynamic and pharmacokinetic sensitivity also have important roles in the generation of arrhythmias. An increase in spatial or temporal dispersion of repolarization and a triangular action-potential configuration have been identified as crucial predictors of proarrhythmia in experimental models. These studies emphasized that sole consideration of the QT interval is not sufficient to assess the proarrhythmic risk. In this Review, we focus on important triggers of proarrhythmia and the underlying electrophysiological mechanisms that can enhance or prevent the development of torsade de pointes.
Collapse
Affiliation(s)
- Gerrit Frommeyer
- Division of Electrophysiology, Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer Strasse 33, D-48149 Münster, Germany
| | - Lars Eckardt
- Division of Electrophysiology, Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer Strasse 33, D-48149 Münster, Germany
| |
Collapse
|
28
|
Gupta T, Khera S, Kolte D, Aronow WS, Iwai S. Antiarrhythmic properties of ranolazine: A review of the current evidence. Int J Cardiol 2015; 187:66-74. [PMID: 25828315 DOI: 10.1016/j.ijcard.2015.03.324] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/20/2015] [Indexed: 12/19/2022]
Abstract
Ranolazine was developed as an antianginal agent and was approved by the Food and Drug Administration in 2006 for use in chronic stable angina pectoris. Experimental and clinical studies have shown that it also has antiarrhythmic properties based on the frequency-dependent blockade of peak sodium channel current (peak INa) and rapidly activating delayed rectifier potassium current (IKr) in the atria and blockade of late phase of the inward sodium current (late INa) in the ventricles. Recent clinical studies have revealed the efficacy of ranolazine in prevention of atrial fibrillation in patients with acute coronary syndromes, prevention as well as conversion of postoperative atrial fibrillation after cardiac surgery, conversion of recent-onset atrial fibrillation and maintenance of sinus rhythm in recurrent atrial fibrillation. Ranolazine has also been shown to reduce ventricular tachycardia and drug-refractory implantable cardioverter defibrillator shocks. The antiarrhythmic effect of ranolazine is preserved in the setting of chronic heart failure and clinical studies have demonstrated its safety in patients with heart failure. This review discusses the available preclinical and clinical data on the antiarrhythmic effects of this novel antianginal agent.
Collapse
Affiliation(s)
- Tanush Gupta
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| | - Sahil Khera
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States.
| | - Dhaval Kolte
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| | - Wilbert S Aronow
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| | - Sei Iwai
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
29
|
Carbon monoxide inhibits inward rectifier potassium channels in cardiomyocytes. Nat Commun 2014; 5:4676. [PMID: 25118981 DOI: 10.1038/ncomms5676] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 07/13/2014] [Indexed: 11/09/2022] Open
Abstract
Reperfusion-induced ventricular fibrillation (VF) severely threatens the lives of post-myocardial infarction patients. Carbon monoxide (CO)--produced by haem oxygenase in cardiomyocytes--has been reported to prevent VF through an unknown mechanism of action. Here, we report that CO prolongs action potential duration (APD) by inhibiting a subset of inward-rectifying potassium (Kir) channels. We show that CO blocks Kir2.2 and Kir2.3 but not Kir2.1 channels in both cardiomyocytes and HEK-293 cells transfected with Kir. CO directly inhibits Kir2.3 by interfering with its interaction with the second messenger phosphatidylinositol (4,5)-bisphosphate (PIP2). As the inhibition of Kir2.2 and Kir2.3 by CO prolongs APD in myocytes, cardiac Kir2.2 and Kir2.3 are promising targets for the prevention of reperfusion-induced VF.
Collapse
|
30
|
Hale SL, Kloner RA. Ranolazine Treatment for Myocardial Infarction? Effects on the Development of Necrosis, Left Ventricular Function and Arrhythmias in Experimental Models. Cardiovasc Drugs Ther 2014; 28:469-75. [DOI: 10.1007/s10557-014-6548-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
31
|
Abstract
Mortality rates attributable to coronary heart disease have declined in recent years, possibly related to changes in clinical presentation patterns and use of proven secondary prevention strategies. Chronic stable angina (CSA) remains prevalent, and the goal of treatment is control of symptoms and reduction in cardiovascular events. Ranolazine is a selective inhibitor of the late sodium current in myocytes with anti-ischemic and metabolic properties. It was approved by the US Food and Drug Administration in 2006 for use in patients with CSA. Multiple, randomized, placebo-controlled trials have shown that ranolazine improves functional capacity and decreases anginal episodes in CSA patients, despite a lack of a significant hemodynamic effect. Ranolazine did not improve cardiovascular mortality or affect incidence of myocardial infarction in the MERLIN (Metabolic Efficiency with Ranolazine for Less Ischemia in Non-ST-Elevation Acute Coronary Syndrome)-TIMI (Thrombolysis In Myocardial Infarction) 36 trial, but significantly decreased the incidence of recurrent angina. More recently, ranolazine has been shown to have beneficial and potent antiarrhythmic effects, both on supraventricular and ventricular tachyarrhythmias, largely due to its inhibition of the late sodium current. Randomized controlled trials testing these effects are underway. Lastly, ranolazine appears to be cost-effective due to its ability to decrease angina-related hospitalizations and improve quality of life.
Collapse
Affiliation(s)
- J Nicolás Codolosa
- Einstein Center for Heart and Vascular Health, Einstein Medical Center, Philadelphia, PA, USA
| | - Subroto Acharjee
- Einstein Center for Heart and Vascular Health, Einstein Medical Center, Philadelphia, PA, USA
| | - Vincent M Figueredo
- Einstein Center for Heart and Vascular Health, Einstein Medical Center, Philadelphia, PA, USA
- Jefferson Medical College, Philadelphia, PA, USA
| |
Collapse
|
32
|
Ranolazine inhibition of hERG potassium channels: drug-pore interactions and reduced potency against inactivation mutants. J Mol Cell Cardiol 2014; 74:220-30. [PMID: 24877995 PMCID: PMC4121676 DOI: 10.1016/j.yjmcc.2014.05.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/14/2014] [Accepted: 05/19/2014] [Indexed: 01/06/2023]
Abstract
The antianginal drug ranolazine, which combines inhibitory actions on rapid and sustained sodium currents with inhibition of the hERG/IKr potassium channel, shows promise as an antiarrhythmic agent. This study investigated the structural basis of hERG block by ranolazine, with lidocaine used as a low potency, structurally similar comparator. Recordings of hERG current (IhERG) were made from cell lines expressing wild-type (WT) or mutant hERG channels. Docking simulations were performed using homology models built on MthK and KvAP templates. In conventional voltage clamp, ranolazine inhibited IhERG with an IC50 of 8.03 μM; peak IhERG during ventricular action potential clamp was inhibited ~ 62% at 10 μM. The IC50 values for ranolazine inhibition of the S620T inactivation deficient and N588K attenuated inactivation mutants were respectively ~ 73-fold and ~ 15-fold that for WT IhERG. Mutations near the bottom of the selectivity filter (V625A, S624A, T623A) exhibited IC50s between ~ 8 and 19-fold that for WT IhERG, whilst the Y652A and F656A S6 mutations had IC50s ~ 22-fold and 53-fold WT controls. Low potency lidocaine was comparatively insensitive to both pore helix and S6 mutations, but was sensitive to direction of K+ flux and particularly to loss of inactivation, with an IC50 for S620T-hERG ~ 49-fold that for WT IhERG. Docking simulations indicated that the larger size of ranolazine gives it potential for a greater range of interactions with hERG pore side chains compared to lidocaine, in particular enabling interaction of its two aromatic groups with side chains of both Y652 and F656. The N588K mutation is responsible for the SQT1 variant of short QT syndrome and our data suggest that ranolazine is unlikely to be effective against IKr/hERG in SQT1 patients. hERG K+ channels regulate cardiac action potential repolarization. The molecular basis of hERG block by ranolazine and structurally related lidocaine was studied. S6 Y652A and F656A mutations affected greatly ranolazine but not lidocaine binding. T623 and S624 residues may directly interact with ranolazine but not lidocaine. N588K and S620T attenuated inactivation mutants had reduced sensitivity to both drugs.
Collapse
|
33
|
|
34
|
Abstract
Myocardial ischemia is a metabolic problem involving reduced delivery of oxygen to cardiac mitochondria, resulting in less ATP formation, acceleration of glycolysis and production of lactate and H+ by the cell. Traditional therapies for ischemia aim at restoring the balance between mitochondrial ATP production and breakdown by reducing the need for ATP via suppression of heart rate, blood pressure and cardiac contractility, or by increasing oxygen delivery via increased myocardial blood flow. Despite optimal treatment with traditional hemodynamically oriented drugs (beta-adrenergic receptor antagonist, Ca2+ channel antagonist and nitrates), many patients continue to suffer from angina. Thus, there is a need for anti-anginal drugs that act directly on cardiomyocytes to lessen the metabolic abnormalities induced by ischemia and reduce the symptoms (chest pain and exercise intolerance). Ranolazine has been demonstrated to improve exercise time to angina or 1 mm of ST-segment depression in a manner similar to currently approved drugs, but without any significant effects on heart rate or blood pressure at rest or during exercise. In two Phase III trials, ranolazine improved exercise tolerance and reduced the frequency of angina attacks in chronic severe angina patients when administered either as monotherapy or on a background of atenolol, amlodinine or diltiazem. At present, ranolazine is under review for US Food and Drug Administration approval and, if approved, it will represent the first drug of its class in the USA.
Collapse
Affiliation(s)
- William C Stanley
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4970, USA.
| |
Collapse
|
35
|
Wang XJ, Wang LL, Fu C, Zhang PH, Wu Y, Ma JH. Ranolazine Attenuates the Enhanced Reverse Na+-Ca2+ Exchange Current via Inhibiting Hypoxia-Increased Late Sodium Current in Ventricular Myocytes. J Pharmacol Sci 2014; 124:365-73. [DOI: 10.1254/jphs.13202fp] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
36
|
Antiarrhythmic effects of simvastatin in canine pulmonary vein sleeve preparations. J Am Coll Cardiol 2013; 57:986-93. [PMID: 21329846 DOI: 10.1016/j.jacc.2010.08.649] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 08/15/2010] [Accepted: 09/23/2010] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The purpose of this study was to determine the electrophysiologic effects of simvastatin in canine pulmonary vein (PV) sleeve preparations. BACKGROUND Ectopic activity arising from the PV plays a prominent role in the development of atrial fibrillation. METHODS Transmembrane action potentials were recorded from canine superfused left superior or inferior PV sleeves using standard microelectrode techniques. Acetylcholine (1 μM), isoproterenol (1 μM), high calcium ([Ca(2+)](o) = 5.4 mM), or a combination was used to induce early afterdepolarizations or delayed afterdepolarizations and triggered activity. Voltage clamp experiments were performed in the left atrium measuring fast and late sodium currents. RESULTS Under steady-state conditions, simvastatin (10 nM, n = 9) induced a small increase in action potential duration measured at 85% repolarization and a significant decrease in action potential amplitude, take-off potential, and maximum rate of rise of action potential upstroke at the fastest rates. The V(max) decreased from 175.1 ± 34 V/s to 151.7 ± 28 V/s and from 142 ± 47 V/s to 97.4 ± 39 V/s at basic cycle lengths of 300 and 200 ms, respectively. Simvastatin (10 to 20 nM) eliminated delayed afterdepolarizations and delayed afterdepolarization-induced triggered activity in 7 of 7 PV sleeve preparations and eliminated or reduced late-phase 3 early afterdepolarizations in 6 of 6 PV sleeve preparations. Simvastatin (20 nM) did not affect late or fast sodium currents measured using voltage clamp techniques. CONCLUSIONS Our data suggest that in addition to its upstream actions to reduce atrial structural remodeling, simvastatin exerts a direct antiarrhythmic effect by suppressing triggers responsible for the genesis of atrial fibrillation.
Collapse
|
37
|
Aistrup GL, Gupta DK, Kelly JE, O'Toole MJ, Nahhas A, Chirayil N, Misener S, Beussink L, Singh N, Ng J, Reddy M, Mongkolrattanothai T, El-Bizri N, Rajamani S, Shryock JC, Belardinelli L, Shah SJ, Wasserstrom JA. Inhibition of the late sodium current slows t-tubule disruption during the progression of hypertensive heart disease in the rat. Am J Physiol Heart Circ Physiol 2013; 305:H1068-79. [PMID: 23873796 DOI: 10.1152/ajpheart.00401.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The treatment of heart failure (HF) is challenging and morbidity and mortality are high. The goal of this study was to determine if inhibition of the late Na(+) current with ranolazine during early hypertensive heart disease might slow or stop disease progression. Spontaneously hypertensive rats (aged 7 mo) were subjected to echocardiographic study and then fed either control chow (CON) or chow containing 0.5% ranolazine (RAN) for 3 mo. Animals were then restudied, and each heart was removed for measurements of t-tubule organization and Ca(2+) transients using confocal microscopy of the intact heart. RAN halted left ventricular hypertrophy as determined from both echocardiographic and cell dimension (length but not width) measurements. RAN reduced the number of myocytes with t-tubule disruption and the proportion of myocytes with defects in intracellular Ca(2+) cycling. RAN also prevented the slowing of the rate of restitution of Ca(2+) release and the increased vulnerability to rate-induced Ca(2+) alternans. Differences between CON- and RAN-treated animals were not a result of different expression levels of voltage-dependent Ca(2+) channel 1.2, sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, ryanodine receptor type 2, Na(+)/Ca(2+) exchanger-1, or voltage-gated Na(+) channel 1.5. Furthermore, myocytes with defective Ca(2+) transients in CON rats showed improved Ca(2+) cycling immediately upon acute exposure to RAN. Increased late Na(+) current likely plays a role in the progression of cardiac hypertrophy, a key pathological step in the development of HF. Early, chronic inhibition of this current slows both hypertrophy and development of ultrastructural and physiological defects associated with the progression to HF.
Collapse
Affiliation(s)
- Gary L Aistrup
- Department of Medicine (Cardiologyand the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The effective treatment of coronary artery disease targets two distinct goals, controlling symptomatic angina and decreasing the adverse events associated with ischemia. Traditional anti-anginal and anti-ischemic drugs function by altering the determinants of myocardial oxygen supply or demand, usually by altering loading conditions, changing the heart rate, or impacting contractility. Blockade of the late inward sodium current, late I(Na), offers another target for the treatment of ischemia. Blockade of late I(Na) reduces the sodium and calcium overload that follows ischemia. This improves myocardial relaxation and reduces left ventricular diastolic stiffness, in turn enhancing myocardial contractility and perfusion. Ranolazine, a late I(Na) inhibitor, has been shown to provide both anti-anginal and anti-ischemic benefits without significant alterations in the heart rate and blood pressure in patients with stable coronary artery disease. When evaluated in patients with acute coronary syndrome, ranolazine has been shown to decrease recurrent ischemia, but not significantly reduce the risk of death or myocardial infarction. This review will address the rationale that inhibition of the late sodium current is beneficial in reducing cardiac dysfunction during ischemia, and discuss the clinical studies supporting the use of ranolazine for its anti-anginal and anti-ischemic effects.
Collapse
|
39
|
Kaye AD, Volpi-Abadie J, Bensler JM, Kaye AM, Diaz JH. QT interval abnormalities: risk factors and perioperative management in long QT syndromes and Torsades de Pointes. J Anesth 2013; 27:575-87. [PMID: 23412014 DOI: 10.1007/s00540-013-1564-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 01/21/2013] [Indexed: 01/08/2023]
Abstract
Electrophysiological abnormalities of the QT interval of the standard electrocardiogram are not uncommon. Congenital long QT syndrome is due to mutations of several possible genes (genotype) that result in prolongation of the corrected QT interval (phenotype). Abnormalities of the QT interval can be acquired and are often drug-induced. Torsades de Pointes (TP) is an arrhythmia that is a result of aberrant repolarization/QT abnormalities. If not recognized and corrected quickly, QT interval abnormalities may precipitate potentially fatal ventricular dysrhythmias. The main mechanism responsible for the development of QT prolongation is blockade of the rapid component of the delayed rectifier potassium current (I kr), encoded for by the human-ether-a-go-go-related gene (hERG). The objectives of this review were (1) to describe the electrical pathophysiology of QT interval abnormalities, (2) to differentiate congenital from acquired QT interval abnormalities, (3) to describe the currently known risk factors for QT interval abnormalities, (4) to identify current drug-induced causes of acquired QT interval abnormalities, and (5) to recommend immediate and effective management strategies to prevent unanticipated dysrhythmias and deaths from QT abnormalities in the perioperative period.
Collapse
Affiliation(s)
- Alan David Kaye
- Department of Anesthesiology, LSU School of Medicine, New Orleans, LA 70112, USA.
| | | | | | | | | |
Collapse
|
40
|
Abstract
The anti-arrhythmic efficacy of the late sodium channel current (late I(Na)) inhibition has been convincingly demonstrated in the ventricles, particularly under conditions of prolonged ventricular repolarization. The value of late I(Na) block in the setting of atrial fibrillation (AF) remains poorly investigated. All sodium channel blockers inhibit both peak and late I(Na) and are generally more potent in inhibiting late vs. early I(Na). Selective late I(Na) block does not prolong the effective refractory period (ERP), a feature common to practically all anti-AF agents. Although the late I(Na) blocker ranolazine has been shown to be effective in suppression of AF, it is noteworthy that at concentrations at which it blocks late I(Na) in the ventricles, it also potently blocks peak I(Na) in the atria, thus causing rate-dependent prolongation of ERP due to development of post-repolarization refractoriness. Late I(Na) inhibition in atria is thought to suppress intracellular calcium (Ca(i))-mediated triggered activity, secondary to a reduction in intracellular sodium (Na(i)). However, agents that block late I(Na) (ranolazine, amiodarone, vernakalant, etc) are also potent atrial-selective peak I(Na) blockers, so that the reduction of Na(i) loading in atrial cells by these agents can be in large part due to the block of peak I(Na). The impact of late I(Na) inhibition is reduced by the abbreviation of the action potential that occurs in AF patients secondary to electrical remodeling. It stands to reason that selective late I(Na) block may contribute more to inhibition of Ca(i)-mediated triggered activity responsible for initiation of AF in clinical pathologies associated with a prolonged atrial APD (such as long QT syndrome). Additional studies are clearly needed to test this hypothesis.
Collapse
|
41
|
Heijman J, Voigt N, Dobrev D. New directions in antiarrhythmic drug therapy for atrial fibrillation. Future Cardiol 2013; 9:71-88. [DOI: 10.2217/fca.12.78] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia and has a significant impact on morbidity and mortality. Current antiarrhythmic drugs for AF suffer from limited safety and efficacy, probably because they were not designed based on specific pathological mechanisms. Recent research has provided important insights into the mechanisms contributing to AF and highlighted several potential novel antiarrhythmic strategies. In this review, we highlight the main pathological mechanisms of AF, discuss traditional and novel aspects of atrial antiarrhythmic drugs in relation to these pathological mechanisms, and present potential novel therapeutic approaches including structure-based modulation of atrial-specific cardiac ion channels, restoring abnormal Ca2+ handling in AF and targeting atrial remodeling.
Collapse
Affiliation(s)
- Jordi Heijman
- Institute of Pharmacology, Medical Faculty Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Niels Voigt
- Institute of Pharmacology, Medical Faculty Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
- Division of Experimental Cardiology, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Dobromir Dobrev
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
42
|
Soliman D, Wang L, Hamming KSC, Yang W, Fatehi M, Carter CC, Clanachan AS, Light PE. Late sodium current inhibition alone with ranolazine is sufficient to reduce ischemia- and cardiac glycoside-induced calcium overload and contractile dysfunction mediated by reverse-mode sodium/calcium exchange. J Pharmacol Exp Ther 2012; 343:325-32. [PMID: 22879384 DOI: 10.1124/jpet.112.196949] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Excessive reverse-mode (RM) sodium/calcium exchanger 1.1 (NCX1.1) activity, resulting from intracellular sodium accumulation caused by reduced Na+/K+-ATPase activity, increased Na-H exchanger 1 activity. The induction of the voltage-gated sodium channel late current component (late INa), is a major pathway for intracellular calcium (Ca2+i) loading in cardiac ischemia-reperfusion (IR) injury and cardiac glycoside toxicity. Inhibition of late INa with the antianginal agent ranolazine is protective in models of IR injury and cardiac glycoside toxicity. However, whether inhibition of late INa alone is sufficient to provide maximal protection or additional inhibition of RM NCX1.1 provides further benefit remains to be determined conclusively. Therefore, the effects of ranolazine were compared with the INa inhibitor lidocaine in models of IR injury and ouabain toxicity, RM NCX1.1-mediated Ca2+ overload, and patch-clamp assays of RM NCX1.1 currents. Ranolazine and lidocaine (10 μM) similarly reduced Ca2+i overload and improved left ventricle work recovery in whole-heart models of IR injury or exposure to ouabain (80 μM). Ranolazine (10 μM), but not lidocaine (10 μM), reduced RM NCX1.1-mediated Ca2+i overload in ventricular myocytes. Furthermore, ranolazine inhibited RM NCX1.1 currents (IC50 1.7 μM), without affecting forward mode currents, revealing that ranolazine has novel RM NCX1.1 inhibitory actions. However, because lidocaine provides similar protection to ranolazine in whole-heart models but does not inhibit RM NCX1.1, we conclude that induction of late INa is upstream of RM NCX1.1 activity and selective inhibition of late INa alone is sufficient to reduce Ca2+i overload and contractile dysfunction in IR injury and cardiac glycoside toxicity.
Collapse
Affiliation(s)
- Daniel Soliman
- Cardiovascular Research Centre, Department of Pharmacology, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, T6G 2E1
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Tzeis S, Andrikopoulos G. Antiarrhythmic properties of ranolazine – from bench to bedside. Expert Opin Investig Drugs 2012; 21:1733-41. [DOI: 10.1517/13543784.2012.716826] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Aidonidis I, Doulas K, Hatziefthimiou A, Tagarakis G, Simopoulos V, Rizos I, Tsilimingas N, Molyvdas PA. Ranolazine-Induced Postrepolarization Refractoriness Suppresses Induction of Atrial Flutter and Fibrillation in Anesthetized Rabbits. J Cardiovasc Pharmacol Ther 2012; 18:94-101. [DOI: 10.1177/1074248412453874] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ranolazine (Ran) is a novel anti-ischemic agent with electrophysiologic properties mainly attributed to the inhibition of late Na+ current and atrial-selective early Na+ current. However, there are only limited data regarding its efficacy and mechanism of action against atrial flutter (Afl) and atrial fibrillation (AF) in intact animals. Therefore, we aimed to investigate the electrophysiologic mechanism of Ran in a rabbit model of inducible atrial tachyarrhythmias elicited by acetylcholine (ACh). Arrhythmias were produced in 19 rabbits by rapid atrial burst pacing during control, after intravenous ACh and after Ran + ACh administration. Recording of right atrial monophasic action potentials (MAPs) and programmed stimulation were utilized to determine the duration of atrial repolarization at various cycle lengths and voltage levels of action potential, including 75% of total MAP duration (MAPD75), effective refractory period (ERP), and postrepolarization refractoriness (PRR = ERP − MAPD75) prior to and after Ran. Control stimulation yielded no arrhythmias or maximal nonsustained runs of Afl/AF. Upon ACh, 17 of 19 rabbits exhibited sustained Afl and AF as well as mixed forms of Afl/AF, while 2 animals revealed none or short runs of nonsustained arrhythmias and were excluded from the study. High-frequency burst pacing during the first 30 minutes after Ran + ACh failed to induce any arrhythmia in 13 of 17 rabbits (76%), while 2 animals displayed sustained Afl/AF and 2 other animals nonsustained Afl/AF. At basic stimulation cycle length of 250 milliseconds, Ran prolonged baseline atrial ERP (80 ± 8 vs 120 ± 9 milliseconds, P < .001) much more than MAPD75 (65 ± 7 vs 85 ± 7 milliseconds, P < .001), leading to atrial PRR which was more pronounced after Ran compared with control measurements (35 ± 11 vs 15 ± 10 milliseconds, P < .001). This in vivo study demonstrates that Ran exerts antiarrhythmic activity by suppressing inducibility of ACh-mediated Afl/AF in intact rabbits. Its action may predominantly be related to a significant increase in atrial PRR, resulting in depressed electrical excitability and impediment of arrhythmia initiation.
Collapse
Affiliation(s)
- Isaac Aidonidis
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Konstantinos Doulas
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Apostolia Hatziefthimiou
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Georgios Tagarakis
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Vassilios Simopoulos
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Ioannis Rizos
- Department of Cardiology, Attikon University Hospital of Athens, Greece
| | - Nikolaos Tsilimingas
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Paschalis-Adam Molyvdas
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| |
Collapse
|
45
|
Long-term nerve excitability changes by persistent Na+ current blocker ranolazine. Neurosci Lett 2012; 524:101-6. [PMID: 22824305 DOI: 10.1016/j.neulet.2012.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 07/04/2012] [Accepted: 07/08/2012] [Indexed: 01/23/2023]
Abstract
The persistent Na(+) current (Na(p)) in peripheral axons plays an important functional role in controlling the axonal excitability. Abnormal Na(p) is believed to contribute to neurodegeneration and neuropathic pain, and thus it is an attractive therapeutic target. To assess the chronic behavior of selective Na(p) blockade, axonal excitability testing was performed in vivo in normal male mice exposed to ranolazine by recording the tail sensory nerve action potentials (SNAPs). Seven days after administering ranolazine i.p. (50mg/kg) daily for 1 week, nerve excitability testing showed decreased strength-duration time constant in the ranolazine group in comparison to the control (P<0.03). This change is explained by the long-term effects of ranolazine on Na(p). Importantly, ranolazine showed no effect on other ion channels that influence axonal excitability. Further study is needed to assess the chronic Na(p) blockade as a useful therapy in peripheral nerve diseases associated with abnormal nerve excitability.
Collapse
|
46
|
Vizzardi E, D'Aloia A, Quinzani F, Bonadei I, Rovetta R, Bontempi L, Curnis A, Dei Cas L. A focus on antiarrhythmic properties of ranolazine. J Cardiovasc Pharmacol Ther 2012; 17:353-6. [PMID: 22492919 DOI: 10.1177/1074248412442000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ranolazine is an antianginal drug that inhibits a number of ion currents that are important for the genesis of transmembrane cardiac action potential. It was initially developed as an antianginal agent but was found to additionally exert antiarrhythmic actions, due to its multichannel-blocking properties. In recent years, several studies about the antiarrhythmic properties of ranolazine were conducted, demonstrating the beneficial effects of this drug in both atrial and ventricular arrhythmias, such as atrial fibrillation, ventricular premature beats, ventricular tachycardia, torsades de pointes, and ventricular fibrillation. Our aim is to briefly review the main points of these studies, most more experimental than clinical.
Collapse
Affiliation(s)
- Enrico Vizzardi
- Department of Applied and Experimental Medicine, Chair of Cardiology University of Brescia, Brescia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Effect of ranolazine on rat intrarenal arteries in vitro. Eur J Pharmacol 2012; 683:211-6. [PMID: 22449375 DOI: 10.1016/j.ejphar.2012.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 03/05/2012] [Accepted: 03/07/2012] [Indexed: 11/21/2022]
Abstract
Ranolazine is mainly used to treat patients with chronic stable angina in clinical practice. However, ranolazine does not lower significantly systemic blood pressure. The direct effect of ranolazine on vascular tone remains unknown. In the present study, we investigated the vascular effects and mechanisms of action of ranolazine in isolated rat intrarenal arteries. Rings of intrarenal arteries were mounted in a small vessel myography using two stainless steel wires for the measurement of isometric tension. L-type Ca²⁺ currents were recorded in isolated single renal arterial smooth muscle cells using patch clamp techniques in whole-cell mode. Ranolazine induced concentration-dependent relaxations in rings contracted with phenylephrine, but ranolazine failed to cause any relaxation in rings pre-contracted by U46619, 5-HT or endothelin-1. Ranolazine also induced relaxations in norepinephrine pre-contracted rings. Yohimbine failed to induce relaxation in rings pre-contracted by norepinephrine. Propranolol did not affect ranolazine-induced relaxation but the relaxant effect of ranolazine was much less than that of prazosin. Ranolazine-induced relaxations were slight but significantly attenuated by endothelial denudation. Partial inhibition was observed in endothelium-intact arteries exposed to a combination of iberiotoxin and apamin. Ranolazine at higher concentration (>30 μM) inhibited Ca²⁺-induced contraction in a noncompetitive manner. Ranolazine reduced L-type Ca²⁺ currents at potentials between -30 and 50 mV in isolated renal artery myocytes. Therefore it can be said that ranolazine has significant α₁-adrenergic receptor and weak calcium channel antagonistic effects in rat intrarenal arteries.
Collapse
|
48
|
Sicouri S, Pourrier M, Gibson JK, Lynch JJ, Antzelevitch C. Comparison of electrophysiological and antiarrhythmic effects of vernakalant, ranolazine, and sotalol in canine pulmonary vein sleeve preparations. Heart Rhythm 2012; 9:422-9. [PMID: 22019863 PMCID: PMC3288874 DOI: 10.1016/j.hrthm.2011.10.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 10/17/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Vernakalant (VER) is a relatively atrial-selective antiarrhythmic drug capable of blocking potassium and sodium currents in a frequency- and voltage-dependent manner. Ranolazine (RAN) is a sodium-channel blocker shown to exert antiarrhythmic effects in pulmonary vein (PV) sleeves. dl-Sotalol (SOT) is a β-blocker commonly used in the rhythm-control treatment of atrial fibrillation. This study evaluated the electrophysiological and antiarrhythmic effects of VER, RAN, and SOT in canine PV sleeve preparations in a blinded fashion. METHODS Transmembrane action potentials were recorded from canine superfused PV sleeve preparations exposed to VER (n = 6), RAN (n = 6), and SOT (n = 6). Delayed afterdepolarizations were induced in the presence of isoproterenol and high-calcium concentrations by periods of rapid pacing. RESULTS In PV sleeves, VER, RAN, and SOT (3-30 μM) produced small (10-15 ms) increases in action potential duration. The effective refractory period, diastolic threshold of excitation, and the shortest S(1)-S(1) cycle length permitting 1:1 activation were significantly increased by VER and RAN in a rate- and concentration-dependent manner. VER and RAN significantly reduced V(max) in a concentration- and rate-dependent manner. SOT did not significantly affect the effective refractory period, V(max), diastolic threshold of excitation, or the shortest S(1)-S(1) cycle length permitting 1:1 activation. All 3 agents (3-30 μM) suppressed delayed afterdepolarization-mediated triggered activity induced by isoproterenol and high calcium. CONCLUSIONS In canine PV sleeves, the effects of VER and RAN were similar and largely characterized by concentration- and rate-dependent depression of sodium-channel-mediated parameters, which were largely unaffected by SOT. All 3 agents demonstrated an ability to effectively suppress delayed afterdepolarization-induced triggers of atrial arrhythmia.
Collapse
|
49
|
Parikh A, Mantravadi R, Kozhevnikov D, Roche MA, Ye Y, Owen LJ, Puglisi JL, Abramson JJ, Salama G. Ranolazine stabilizes cardiac ryanodine receptors: a novel mechanism for the suppression of early afterdepolarization and torsades de pointes in long QT type 2. Heart Rhythm 2012; 9:953-60. [PMID: 22245792 DOI: 10.1016/j.hrthm.2012.01.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ranolazine (Ran) is known to inhibit multiple targets, including the late Na(+)current, the rapid delayed rectifying K(+)current, the L-type Ca(2+)current, and fatty acid metabolism. Functionally, Ran suppresses early afterdepolarization (EADs) and torsades de pointes (TdP) in drug-induced long QT type 2 (LQT2) presumably by decreasing intracellular [Na(+)](i) and Ca(2+)overload. However, simulations of EADs in LQT2 failed to predict their suppression by Ran. OBJECTIVE To elucidate the mechanism(s) whereby Ran alters cardiac action potentials (APs) and cytosolic Ca(2+)transients and suppresses EADs and TdP in LQT2. METHODS The known effects of Ran were included in simulations (Shannon and Mahajan models) of rabbit ventricular APs and Ca(2+)transients in control and LQT2 models and compared with experimental optical mapping data from Langendorff rabbit hearts treated with E4031 (0.5 μM) to block the rapid delayed rectifying K(+)current. Direct effects of Ran on cardiac ryanodine receptors (RyR2) were investigated in single channels and changes in Ca(2+)-dependent high-affinity ryanodine binding. RESULTS Ran (10 μM) alone prolonged action potential durations (206 ± 4.6 to 240 ± 7.8 ms; P <0.05); E4031 prolonged action potential durations (204 ± 6 to 546 ± 35 ms; P <0.05) and elicited EADs and TdP that were suppressed by Ran (10 μM; n = 7 of 7 hearts). Simulations (Shannon but not Mahajan model) closely reproduced experimental data except for EAD suppression by Ran. Ran reduced open probability (P(o)) of RyR2 (half maximal inhibitory concentration = 10 ± 3 μM; n = 7) in bilayers and shifted half maximal effective concentration for Ca(2+)-dependent ryanodine binding from 0.42 ± 0.02 to 0.64 ± 0.02 μM with 30 μM Ran. CONCLUSIONS Ran reduces P(o) of RyR2, desensitizes Ca(2+)-dependent RyR2 activation, and inhibits Ca(i) oscillations, which represents a novel mechanism for its suppression of EADs and TdP.
Collapse
Affiliation(s)
- Ashish Parikh
- Department of Bioengineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim HJ, Ahn HS, Choi JS, Choi BH, Hahn SJ. Effects of ranolazine on cloned cardiac kv4.3 potassium channels. J Pharmacol Exp Ther 2011; 339:952-8. [PMID: 21940646 DOI: 10.1124/jpet.111.184176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
The effects of ranolazine, an antianginal drug, on potassium channel Kv4.3 were examined by using the whole-cell patch-clamp technique. Ranolazine inhibited the peak amplitude of Kv4.3 in a reversible, concentration-dependent manner with an IC(50) of 128.31 μM. The activation kinetics were not significantly affected by ranolazine at concentrations up to 100 μM. Applications of 10 and 30 μM ranolazine had no effect on the fast and slow inactivation of Kv4.3. However, at concentrations of 100 and 300 μM ranolazine caused a significant decrease in the rate of fast inactivation, and at a concentration of 300 μM it caused a significant decrease in the rate of slow inactivation, resulting in a crossover of the current traces during depolarization. The Kv4.3 inhibition by ranolazine increased steeply between -20 and +20 mV. In the full activation voltage range, however, no voltage-dependent inhibition was found. Ranolazine shifted the voltage dependence of the steady-state inactivation of Kv4.3 in the hyperpolarizing direction in a concentration-dependent manner. The apparent dissociation constant (K(i)) for ranolazine for interacting with the inactivated state of Kv4.3 was calculated to be 0.32 μM. Ranolazine produced little use-dependent inhibition at frequencies of 1 and 2 Hz. Ranolazine did not affect the time course of recovery from the inactivation of Kv4.3. The results indicated that ranolazine inhibited Kv4.3 and exhibited a low affinity for Kv4.3 channels in the closed state but a much higher affinity for Kv4.3 channels in the inactivated state.
Collapse
Affiliation(s)
- Hee Jae Kim
- Department of Physiology, Medical Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul 137-701, Korea
| | | | | | | | | |
Collapse
|