1
|
Ellermann C, Mengel C, Wolfes J, Wegner FK, Rath B, Köbe J, Eckardt L, Frommeyer G. Sacubitril Does Not Exert Proarrhythmic Effects in Combination with Different Antiarrhythmic Drugs. Pharmaceuticals (Basel) 2025; 18:230. [PMID: 40006043 PMCID: PMC11859141 DOI: 10.3390/ph18020230] [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: 12/30/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Previous studies suggest a direct effect of sacubitril on cardiac electrophysiology and indicate potential arrhythmic interactions between sacubitril and antiarrhythmic drugs. Therefore, the aim of this study was to explore the electrophysiologic effects of combining sacubitril with the antiarrhythmic drugs d,l-sotalol and mexiletine in isolated hearts. Methods and results: A total of 25 rabbit hearts were perfused using a Langendorff setup. Following baseline data collection, hearts were treated with mexiletine (25 µM, 13 hearts) or d,l-sotalol (100 µM, 12 hearts). Monophasic action potential demonstrated an abbreviation of action potential duration (APD90) after administration of mexiletine. Spatial dispersion of repolarization remained unchanged after mexiletine treatment, whereas effective refractory periods (ERP) were significantly prolonged. D,l-sotalol prolonged cardiac repolarization and amplified spatial dispersion. Further infusion of sacubitril (5 µM) led to a significant reduction in APD90 and ERP in the mexiletine group. In the d,l-sotalol group, additional administration of sacubitril shortened cardiac repolarization duration without affecting spatial dispersion. No proarrhythmic effect was observed after mexiletine treatment as assessed by a predefined pacing protocol. Additional sacubitril treatment did not increase ventricular vulnerability. When potassium concentration was reduced, 30 episodes of torsade de pointes tachycardia occurred after d,l-sotalol treatment. Additional sacubitril treatment significantly suppressed torsade de pointes tachycardia (eight episodes) in the d,l-sotalol-group. Conclusions: In class IB- and class III-pretreated hearts, sacubitril shortened refractory periods and cardiac repolarization duration. The combination of sacubitril with the antiarrhythmic drugs d,l-sotalol and mexiletine demonstrates a safe electrophysiologic profile and sacubitril reduces the occurrence of class III-related proarrhythmia, i.e., torsade de pointes tachycardia.
Collapse
Affiliation(s)
- Christian Ellermann
- Department of Cardiology II (Electrophysiology), University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Alhourani N, Wolfes J, Könemann H, Ellermann C, Frommeyer G, Güner F, Lange PS, Reinke F, Köbe J, Eckardt L. Relevance of mexiletine in the era of evolving antiarrhythmic therapy of ventricular arrhythmias. Clin Res Cardiol 2024; 113:791-800. [PMID: 38353682 PMCID: PMC11108884 DOI: 10.1007/s00392-024-02383-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/19/2024] [Indexed: 05/22/2024]
Abstract
Despite impressive developments in the field of ventricular arrhythmias, there is still a relevant number of patients with ventricular arrhythmias who require antiarrhythmic drug therapy and may, e.g., in otherwise drug and/or ablation refractory situations, benefit from agents known for decades, such as mexiletine. Through its capability of blocking fast sodium channels in cardiomyocytes, it has played a minor to moderate antiarrhythmic role throughout the recent decades. Nevertheless, certain patients with structural heart disease suffering from drug-refractory, i.e., mainly amiodarone refractory ventricular arrhythmias, as well as those with selected forms of congenital long QT syndrome (LQTS) may nowadays still benefit from mexiletine. Here, we outline mexiletine's cellular and clinical electrophysiological properties. In addition, the application of mexiletine may be accompanied by various potential side effects, e.g., nausea and tremor, and is limited by several drug-drug interactions. Thus, we shed light on the current therapeutic role of mexiletine for therapy of ventricular arrhythmias and discuss clinically relevant aspects of its indications based on current evidence.
Collapse
Affiliation(s)
- Nawar Alhourani
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany.
| | - Julian Wolfes
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Hilke Könemann
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Christian Ellermann
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Gerrit Frommeyer
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Fatih Güner
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Philipp Sebastian Lange
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Florian Reinke
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Julia Köbe
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Lars Eckardt
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
3
|
Ali H, Cristiano E, Lupo P, Foresti S, DE Ambroggi G, DE Lucia C, Turturiello D, Paganini EM, Bessi R, Farghaly AA, Nicolì L, Cappato R. Oral mexiletine for ventricular tachyarrhythmias treatment in implantable cardioverter-defibrillator patients: a systematic review of the literature. Minerva Cardiol Angiol 2023; 71:622-630. [PMID: 36305779 DOI: 10.23736/s2724-5683.22.06176-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
INTRODUCTION To evaluate the clinical outcomes of oral mexiletine (oMXT) to treat ventricular tachyarrhythmias (VTAs) in the era of implantable cardioverter-defibrillator (ICD) technology. EVIDENCE ACQUISITION A systematic search was conducted using PubMed, Embase and Cochrane databases following the PRISMA guidelines to collect literature data reporting oMXT efficacy and safety outcomes in treating VTAs in ICD recipients. EVIDENCE SYNTHESIS Final analysis included four studies accounting for a total of 91 patients with recurrent VTAs treated with oMXT. Amiodarone therapy was initially attempted in most patients (91.2%), while catheter ablation was performed in one-third of patients. VTA recurrences were observed in 55/91 patients (60.4%) during oMXT treatment compared to 91/91 (100%) before treatment (P<0.001). Appropriate therapies occurred in 55/88 ICD patients (62.5%) during oMXT treatment compared to 80/88 (90.9%) before treatment (P<0.001). After oMXT introduction, there was a significant reduction of the individual burden of VTA episodes and appropriate ICD therapies per patient, showing Hedges'g values of -1.103 (P=0.002) and -1.474 (P=0.008), respectively. Safety analysis showed a sample-weighted overall side-effect rate of 30%, while 21% of patients required drug reduction or discontinuation. Aggregated meta-regression analysis of the included studies and remote literature revealed a linear correlation between oMXT dosage and the overall side effects rate (r2 = 0.48; P=0.014). CONCLUSIONS Oral mexiletine provides an adjunctive treatment to manage VTAs and reduces appropriate therapies in ICD patients with moderate efficacy and acceptable safety profiles. These observations await confirmation through randomised clinical trials.
Collapse
Affiliation(s)
- Hussam Ali
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy -
| | - Ernesto Cristiano
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University, Rome, Italy
| | - Pierpaolo Lupo
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Sara Foresti
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Guido DE Ambroggi
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Carmine DE Lucia
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Dario Turturiello
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Edoardo M Paganini
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Riccardo Bessi
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Ahmad A Farghaly
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
- Department of Cardiovascular Medicine, Faculty of Medicine, University of Assiut, Assiut, Egypt
| | - Leoluca Nicolì
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Riccardo Cappato
- Arrhythmia and Electrophysiology Center, IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| |
Collapse
|
4
|
Nasilli G, Yiangou L, Palandri C, Cerbai E, Davis RP, Verkerk AO, Casini S, Remme CA. Beneficial effects of chronic mexiletine treatment in a human model of SCN5A overlap syndrome. Europace 2023; 25:euad154. [PMID: 37369559 PMCID: PMC10299896 DOI: 10.1093/europace/euad154] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS SCN5A mutations are associated with various cardiac phenotypes, including long QT syndrome type 3 (LQT3), Brugada syndrome (BrS), and cardiac conduction disease (CCD). Certain mutations, such as SCN5A-1795insD, lead to an overlap syndrome, with patients exhibiting both features of BrS/CCD [decreased sodium current (INa)] and LQT3 (increased late INa). The sodium channel blocker mexiletine may acutely decrease LQT3-associated late INa and chronically increase peak INa associated with SCN5A loss-of-function mutations. However, most studies have so far employed heterologous expression systems and high mexiletine concentrations. We here investigated the effects of a therapeutic dose of mexiletine on the mixed phenotype associated with the SCN5A-1795insD mutation in HEK293A cells and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS AND RESULTS To assess only the chronic effects on trafficking, HEK293A cells transfected with wild-type (WT) SCN5A or SCN5A-1795insD were incubated for 48 h with 10 µm mexiletine followed by wash-out, which resulted in an increased peak INa for both SCN5A-WT and SCN5A-1795insD and an increased late INa for SCN5A-1795insD. Acute re-exposure of HEK293A cells to 10 µm mexiletine did not impact on peak INa but significantly decreased SCN5A-1795insD late INa. Chronic incubation of SCN5A-1795insD hiPSC-CMs with mexiletine followed by wash-out increased peak INa, action potential (AP) upstroke velocity, and AP duration. Acute re-exposure did not impact on peak INa or AP upstroke velocity, but significantly decreased AP duration. CONCLUSION These findings demonstrate for the first time the therapeutic benefit of mexiletine in a human cardiomyocyte model of SCN5A overlap syndrome.
Collapse
Affiliation(s)
- Giovanna Nasilli
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Chiara Palandri
- Department NeuroFarBa, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| | - Elisabetta Cerbai
- Department NeuroFarBa, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Waszkielewicz A, Marona H, Pańczyk-Straszak K, Filipek B, Rapacz A, Sałat K, Kubacka M, Cios A, Fedak F, Walczak M, Hubicka U, Kwiecień A, Żuromska-Witek B, Szafrański PW, Koczurkiewicz-Adamczyk P, Pękala E, Przejczowska-Pomierny K, Pociecha K, Wyska E. KM-408, a novel phenoxyalkyl derivative as a potential anticonvulsant and analgesic compound for the treatment of neuropathic pain. Pharmacol Rep 2023; 75:128-165. [PMID: 36401763 PMCID: PMC9889419 DOI: 10.1007/s43440-022-00431-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Epilepsy frequently coexists with neuropathic pain. Our approach is based on the search for active compounds with multitarget profiles beneficial in terms of potential side effects and on the implementation of screening for potential multidirectional central activity. METHODS Compounds were synthesized by means of chemical synthesis. After antiseizure and neurotoxicity screening in vivo, KM-408 and its enantiomers were chosen for analgesic activity evaluations. Further safety studies included acute toxicity in mice, the effect on normal electrocardiogram and on blood pressure in rats, whole body plethysmography in rats, and in vitro and biochemical assays. Pharmacokinetics has been studied in rats after iv and po administration. Metabolism has been studied in vivo in rat serum and urine. Radioligand binding studies were performed as part of the mechanism of action investigation. RESULTS Selected results for KM-408: Ki sigma = 7.2*10-8; Ki 5-HT1A = 8.0*10-7; ED50 MES (mice, ip) = 13.3 mg/kg; formalin test (I phase, mice, ip)-active at 30 mg/kg; SNL (rats, ip)-active at 6 mg/kg; STZ-induced pain (mice, ip)-active at 1 mg/kg (von Frey) and 10 mg/kg (hot plate); hot plate test (mice, ip)-active at 30 mg/kg; ED50 capsaicin test (mice, ip) = 18.99 mg/kg; tail immersion test (mice)-active at 0.5%; corneal anesthesia (guinea pigs)-active at 0.125%; infiltration anesthesia (guinea pigs)-active at 0.125%. CONCLUSIONS Within the presented study a novel compound, R,S-2-((2-(2-chloro-6-methylphenoxy)ethyl)amino)butan-1-ol hydrochloride (KM-408) with dual antiseizure and analgesic activity has been developed for potential use in neuropathic pain treatment.
Collapse
Affiliation(s)
- Anna Waszkielewicz
- Department of Bioorganic Chemistry, Chair of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Henryk Marona
- Department of Bioorganic Chemistry, Chair of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Katarzyna Pańczyk-Straszak
- Department of Bioorganic Chemistry, Chair of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Barbara Filipek
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Rapacz
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Monika Kubacka
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Agnieszka Cios
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Filip Fedak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzyńskiego 14, 30-348 Kraków, Poland
| | - Maria Walczak
- Chair and Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Urszula Hubicka
- Chair of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Kwiecień
- Chair of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Barbara Żuromska-Witek
- Chair of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Przemysław W. Szafrański
- Chair of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Paulina Koczurkiewicz-Adamczyk
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Katarzyna Przejczowska-Pomierny
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Krzysztof Pociecha
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
6
|
Farkowski MM, Karlinski M, Pytkowski M, de Asmundis C, Lewandowski M, Mugnai G, Conte G, Marijon E, Anic A, Boveda S, Providencia R. Mexiletine for recurrent ventricular tachycardia in adult patients with structural heart disease and implantable cardioverter defibrillator: an EHRA systematic review. Europace 2022; 24:1504-1511. [DOI: 10.1093/europace/euac101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
The aim of the study was to systematically review evidence on the effectiveness and safety of oral mexiletine administered in monotherapy or in combination with other antiarrhythmic drugs for recurrent ventricular arrhythmia (ventricular tachycardia/ventricular fibrillation, VT/VF) in adult patients with structural heart disease (SHD) and implantable cardioverter defibrillators (ICDs). We systematically searched MEDLINE, Embase, and CENTRAL databases from inception to 27 August 2021 for prospective and retrospective studies investigating mexiletine in the target population. The main outcome was the reduction of ICD therapy. The main safety outcome was the presence of any serious adverse events (SAEs) leading to mexiletine discontinuation. Study quality was assessed using the Cochrane risk of bias tool or the Newcastle–Ottawa scale. Four studies comprising 86 mexiletine recipients were included in the review. We also obtained individual data of 50 patients from two studies. Ischaemic cardiomyopathy (ICM) was present in 86% of patients. The quality of included studies was moderate/low. A narrative review was undertaken as studies varied widely in terms of study population and treatment. Across studies, mexiletine treatment (with or without amiodarone) seemed to consistently reduce the number of ICD therapies especially in a population where catheter ablation (CA) was unsuccessful or contraindicated. In ICM patients deemed eligible for CA, mexiletine seemed to be inferior to CA. Mexiletine was discontinued in 14% of cases, mainly for gastrointestinal or neurological SAE. Mexiletine seems to be an option for the long-term treatment of recurrent VT/VF in adult patients with SHD, especially ICM, and ICD in whom CA was unsuccessful or not suitable.
Collapse
Affiliation(s)
- Michal Miroslaw Farkowski
- II Department of Heart Arrhythmia, National Institute of Cardiology , Alpejska 42, 04-628 Warsaw , Poland
| | - Michal Karlinski
- II Department of Neurology, Institute of Psychiatry and Neurology , Warsaw , Poland
| | - Mariusz Pytkowski
- II Department of Heart Arrhythmia, National Institute of Cardiology , Alpejska 42, 04-628 Warsaw , Poland
| | - Carlo de Asmundis
- Heart Rhythm Management Centre, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel , Brussels , Belgium
| | - Michal Lewandowski
- II Department of Heart Arrhythmia, National Institute of Cardiology , Alpejska 42, 04-628 Warsaw , Poland
| | - Giacomo Mugnai
- Division of Cardiology, West Vicenza General Hospitals , Arzignano (Vicenza) , Italy
- Division of Cardiology, Department of Medicine, University Hospital of Verona , Verona , Italy
| | - Giulio Conte
- Cardiology Department, Cardiocentro Ticino Institute , Lugano , Switzerland
| | - Eloi Marijon
- Département de Cardiologie, Université Paris-Descartes, Hôpital Européen Georges Pompidou , Paris , France
| | - Ante Anic
- Department of Cardiology, University Clinical Hospital Split , Split , Croatia
| | - Serge Boveda
- Heart Rhythm Management Centre, Universitair Ziekenhuis Brussel—Vrije Universiteit Brussel , Brussels , Belgium
- Heart Rhythm Management Department, Clinique Pasteur , Toulouse , France
| | - Rui Providencia
- St Bartholomew’s Hospital, Barts Heart Centre, Barts Health NHS Trust , London , UK
- Institute of Health Informatics, University College of London , London , UK
| |
Collapse
|
7
|
Watkins B, Schuster HM, Gerwin L, Schoser B, Kröger S. The effect of methocarbamol and mexiletine on murine muscle spindle function. Muscle Nerve 2022; 66:96-105. [PMID: 35373353 DOI: 10.1002/mus.27546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION/AIMS The muscle relaxant methocarbamol and the antimyotonic drug mexiletine are widely used for the treatment of muscle spasms, myotonia, and pain syndromes. To determine whether these drugs affect muscle spindle function, we studied their effect on the resting discharge and on stretch-induced action potential frequencies of proprioceptive afferent neurons. METHODS Single unit action potential frequencies of proprioceptive afferents from muscle spindles in the murine extensor digitorum longus muscle of adult C57BL/6J mice were recorded under resting conditions and during ramp-and-hold stretches. Maximal tetanic force of the same muscle after direct stimulation was determined. High-resolution confocal microscopy analysis was performed to determine the distribution of Nav 1.4 channels, a potential target for both drugs. RESULTS Methocarbamol and mexiletine inhibited the muscle spindle resting discharge in a dose-dependent manner with IC50 values around 300 μM and 6 μM, respectively. With increasing concentrations of both drugs, the response to stretch was also affected, with the static sensitivity first followed by the dynamic sensitivity. At high concentrations, both drugs completely blocked muscle spindle afferent output. Both drugs also reversibly reduced the specific force of the extensor digitorum longus muscle after tetanic stimulation. Finally, we present evidence for the presence and specific localization of the voltage-gated sodium channel Nav 1.4 in intrafusal fibers. DISCUSSION In this study we demonstrate that both muscle relaxants affect muscle spindle function, suggesting impaired proprioception as a potential side effect of both drugs. Moreover, our results provide additional evidence of a peripheral activity of methocarbamol and mexiletine.
Collapse
Affiliation(s)
- Bridgette Watkins
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| | - Hedwig M Schuster
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| | - Laura Gerwin
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| | - Benedikt Schoser
- Department of Neurology, LMU Klinikum, Friedrich-Baur-Institute, Ludwig-Maximilians-University, Munich, Germany
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| |
Collapse
|
8
|
Abstract
Background:
Mexiletine belongs to the β-amino-aryl-ether group of pharmaceutical and applied in the diagnosis of antiarrhythmics, allodynia, and myotonic disorders. In its chemical structure, it possesses a chiral center and practiced in the form of the racemic mixture. The production and accessibility of mexiletine have accompanied with a meaningful development in awareness of its pharmacologic actions. But in clinical arrhythmias and binding experiments on cardiac sodium channels, the (R)-enantiomer of mexiletine is more potent than the (S)-enantiomer. Also, (S)-enantiomer is further active in the diagnosis of allodynia than the (R)-enantiomer.
Methods:
During the last two decades, chromatographic techniques such as HPLC, and GC coupled with mass spectrometry or field ionization detector was used for the stereoselective analysis of MEX enantiomers.
Results:
The direct enantioresolution deal with the use of chiral stationary phases (CSPs) with or no pre-derivatization which depend on a chromophoric entity in the racemates whereas indirect HPLC process involved the use of chiral derivatization reagents (CDR) for the synthesis of diastereomeric derivatives of racemates. Different techniques have their strengths and weaknesses.
Conclusion:
Regulation of enantiomeric purity and estimation of particular enantiomers of drug molecules stays an essential topic for therapeutic, diagnostic, and regulatory uses and to promote a precise assessment of the hazards to human health by false enantiomers. This review aims to offer a systematic survey of the analytical methods (chromatography based) used in the enantioselective analysis of MEX developed in the last two decades (the year 2000 onwards).
Collapse
|
9
|
St Denis JD, Hall RJ, Murray CW, Heightman TD, Rees DC. Fragment-based drug discovery: opportunities for organic synthesis. RSC Med Chem 2020; 12:321-329. [PMID: 34041484 PMCID: PMC8130625 DOI: 10.1039/d0md00375a] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022] Open
Abstract
This Review describes the increasing demand for organic synthesis to facilitate fragment-based drug discovery (FBDD), focusing on polar, unprotected fragments. In FBDD, X-ray crystal structures are used to design target molecules for synthesis with new groups added onto a fragment via specific growth vectors. This requires challenging synthesis which slows down drug discovery, and some fragments are not progressed into optimisation due to synthetic intractability. We have evaluated the output from Astex's fragment screenings for a number of programs, including urokinase-type plasminogen activator, hematopoietic prostaglandin D2 synthase, and hepatitis C virus NS3 protease-helicase, and identified fragments that were not elaborated due, in part, to a lack of commercially available analogues and/or suitable synthetic methodology. This represents an opportunity for the development of new synthetic research to enable rapid access to novel chemical space and fragment optimisation.
Collapse
Affiliation(s)
| | - Richard J Hall
- Astex Pharmaceuticals 436 Cambridge Science Park Cambridge CB4 0QA UK
| | | | - Tom D Heightman
- Astex Pharmaceuticals 436 Cambridge Science Park Cambridge CB4 0QA UK
| | - David C Rees
- Astex Pharmaceuticals 436 Cambridge Science Park Cambridge CB4 0QA UK
| |
Collapse
|
10
|
Brooks EK, Schweitzer D, Robinson HL. A case of paramyotonia congenita in pregnancy. Obstet Med 2020; 13:192-194. [PMID: 33343696 PMCID: PMC7726174 DOI: 10.1177/1753495x18816171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 11/05/2018] [Indexed: 11/17/2022] Open
Abstract
Paramyotonia congenita is a rare autosomal dominant non-dystrophic myopathy caused by mutations in the SNC4A gene, which encodes for the voltage-gated sodium channel in skeletal muscle. Symptom onset is typically during early childhood and is characterised by myotonia followed by flaccid paralysis or weakness, usually exacerbated by repeated muscle contractions or cold temperatures. Pregnancy has been reported to increase symptoms of myotonia; however, there is limited information in the literature regarding the possible effects of paramyotonia congenita on pregnancy and labour. We present a successful case of a 20-year-old primigravida with confirmed paramyotonia congenita and review the literature regarding paramyotonia congenita during pregnancy.
Collapse
Affiliation(s)
- EK Brooks
- Department of Medicine, Ipswich Hospital, Ipswich, Queensland, Australia
| | - D Schweitzer
- Department of Neurology, Mater Hospital, Brisbane, Queensland, Australia
| | - HL Robinson
- Department of Medicine, Ipswich Hospital, Ipswich, Queensland, Australia
- Department of Obstetric Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Pinto DP, Coutinho DDS, Carvalho KIMD, Ferrero MR, Silva LVD, Silveira GPE, Silva DMD, Araújo JFG, Silva ACA, Pereira HM, Fonseca LBD, Faria RX, Souza MVND, Silva ETD, Santos-Filho OA, Costa JCSD, Amendoeira FC, Martins MA. Pharmacological profiling of JME-173, a novel mexiletine derivative combining dual anti-inflammatory/anti-spasmodic functions and limited action in Na + channels. Eur J Pharmacol 2020; 885:173367. [PMID: 32750364 DOI: 10.1016/j.ejphar.2020.173367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
Existing evidence suggests that the local anaesthetic mexiletine can be beneficial for patients with asthma. However, caution is required since anaesthesia of the airways inhibits protective bronchodilator neuronal reflexes, limiting applications in conditions of hyperirritable airways. Here, we describe the synthesis of a new series of mexiletine analogues, which were screened for reduced activity in Na+ channels and improved smooth muscle relaxant effects, that were evaluated using the patch-clamp technique and an isolated tracheal organ bath, respectively. JME-173 (1-(4-bromo-3,5-dimethylphenoxy)propan-2-amine) was the most effective among the four mexiletine analogues investigated. JME-173 was then studied in vivo using a murine model of lung inflammation induced by cigarette smoke (CS) and in vitro using neutrophil chemotaxis and mast cell degranulation assays. Finally, the JME-173 pharmacokinetic profile was assessed using HPLC-MS/MS bioanalytical method. JME-173 directly inhibited IL-8 (CXCL8)- and FMLP-induced human neutrophil chemotaxis and allergen-induced mast cell degranulation. After oral administration 1 h before CS exposure, JME-173 (50 mg/kg) strongly reduced the increased number of macrophages and neutrophils recovered in the bronchoalveolar effluent without altering lymphocyte counts. Pharmacokinetic experiments of JME-173 (10 mg/kg, orally) showed values of maximum concentration (Cmax), maximum time (Tmax), area under the blood concentration-time curve (AUC0-t) and area under the blood concentration-time curve from 0-Inf (AUC0-inf) of 163.3 ± 38.3 ng/mL, 1.2 ± 0.3 h, 729.4 ± 118.3 ng*h/ml and 868.9 ± 117.1 ng*h/ml (means ± S.E.M.), respectively. Collectively, these findings suggest that JME-173 has the potential to be an effective oral treatment for diseases associated with bronchoconstriction and inflammation.
Collapse
Affiliation(s)
- Douglas Pereira Pinto
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Diego de Sá Coutinho
- Laboratory of Inflammation, Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Maximiliano R Ferrero
- Laboratory of Inflammation, Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Letícia Vallim da Silva
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Diego Medeiros da Silva
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - João Felipe Garcia Araújo
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Aline C A Silva
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Heliana Martins Pereira
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Laís Bastos da Fonseca
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Robson Xavier Faria
- Laboratory of Toxoplasmosis and Other Protozoans, Oswaldo Cruz Institute, Brazil
| | - Marcus Vinicius Nora de Souza
- Laboratory of Organic Synthesis, Institute of Technology in Drugs, Farmanguinhos - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Emerson Teixeira da Silva
- Laboratory of Organic Synthesis, Institute of Technology in Drugs, Farmanguinhos - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Osvaldo Andrade Santos-Filho
- Laboratory of Molecular Modeling and Computational Structural Biology, Instituto de Pesquisas de Produtos Naturais - Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fábio Coelho Amendoeira
- Department of Pharmacology and Toxicology, National Institute of Quality Control in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marco Aurélio Martins
- Laboratory of Inflammation, Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
12
|
Carvalho KIM, Coutinho DDS, Joca HC, Miranda AS, Cruz JDS, Silva ET, Souza MVN, Faria RX, Silva PMRE, Costa JCS, Martins MA. Anti-Bronchospasmodic Effect of JME-173, a Novel Mexiletine Analog Endowed With Highly Attenuated Anesthetic Activity. Front Pharmacol 2020; 11:1159. [PMID: 32903732 PMCID: PMC7438868 DOI: 10.3389/fphar.2020.01159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 07/16/2020] [Indexed: 01/16/2023] Open
Abstract
Local anesthetics (LAs), such as lidocaine and mexiletine, inhibit bronchoconstriction in asthmatics, but adverse effects limit their use for this specific clinical application. In this study, we describe the anti-spasmodic properties of the mexiletine analog 2-(2-aminopropoxy)-3,5-dimethyl, 4-Br-benzene (JME-173), which was synthesized and screened for inducing reduced activity on Na+ channels. The effectiveness of JME-173 was assessed using rat tracheal rings, a GH3 cell line and mouse cardiomyocytes to access changes in smooth muscle contraction, and Na+, and Ca++ionic currents, respectively. Bronchospasm and airway hyper-reactivity (AHR) were studied using whole-body barometric plethysmography in A/J mice. We observed that the potency of JME-173 was 653-fold lower than mexiletine in inhibiting Na+ currents, but 12-fold higher in inhibiting L-type Ca++ currents. JME-173 was also more potent than mexiletine in inhibiting tracheal contraction by carbachol, allergen, extracellular Ca++, or sodium orthovanadate provocations. The effect of JME-173 on carbachol-induced tracheal contraction remained unaltered under conditions of de-epithelized rings, β2-receptor blockade or adenylate cyclase inhibition. When orally administered, JME-173 and theophylline inhibited methacholine-induced bronchospasm at time points of 1 and 3 h post-treatment, while only JME-173 remained active for at least 6 h. In addition, JME-173 also inhibited AHR in a mouse model of lipopolysaccharide (LPS)-induced lung inflammation. Thus, the mexiletine analog JME-173 shows highly attenuated activity on Na+ channels and optimized anti-spasmodic properties, in a mechanism that is at least in part mediated by regulation of Ca++ inflow toward the cytosol. Thus, JME-173 is a promising alternative for the treatment of clinical conditions marked by life-threatening bronchoconstriction.
Collapse
Affiliation(s)
| | | | - Humberto Cavalcante Joca
- Laboratory of Excitable Membranes and Cardiovascular Biology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Artur Santos Miranda
- Laboratory of Excitable Membranes and Cardiovascular Biology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jader Dos Santos Cruz
- Laboratory of Excitable Membranes and Cardiovascular Biology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Robson Xavier Faria
- Laboratory of Toxoplasmosis and Other Protozoans, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
13
|
Hesketh LM, Wilder CDE, Ranadive NN, Lytra G, Qazimi P, Munro JS, Ahdi N, Curtis MJ. Characterisation of mexiletine's translational therapeutic index for suppression of ischaemia-induced ventricular fibrillation in the rat isolated heart. Sci Rep 2020; 10:8397. [PMID: 32439959 PMCID: PMC7242333 DOI: 10.1038/s41598-020-65190-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/24/2020] [Indexed: 11/09/2022] Open
Abstract
The 'translational therapeutic index' (TTI) is a drug's ratio of nonclinical threshold dose (or concentration) for significant benefit versus threshold for adversity. In early nonclinical research, discovery and safety studies are normally undertaken separately. Our aim was to evaluate a novel integrated approach for generating a TTI for drugs intended for prevention of ischaemia-induced ventricular fibrillation (VF). We templated the current best available class 1b antiarrhythmic, mexiletine, using the rat Langendorff preparation. Mexiletine's beneficial effects on the incidence of VF caused by 120 min regional ischaemia were contrasted with its concurrent adverse effects (on several variables) in the same hearts, to generate a TTI. Mexiletine 0.1 and 0.5 µM had no adverse effects, but did not reduce VF incidence. Mexiletine 1 µM reduced VF incidence to 0% but had adverse effects on atrioventricular conduction and ventricular repolarization. Separate studies undertaken using an intraventricular balloon revealed no detrimental effects of mexiletine (1 and 5 µM) on mechanical function, or any benefit against reperfusion-related dysfunction. Mexiletine's TTI was found to be less than two, which accords with its clinical therapeutic index. Although non-cardiac adversity, identifiable from additional in vivo studies, may reduce the TTI further, it cannot increase it. Our experimental approach represents a useful early-stage integrated risk/benefit method that, when TTI is found to be low, would eliminate unsuitable class 1b drugs prior to next stage in vivo work, with mexiletine's TTI defining the gold standard that would need to be bettered.
Collapse
Affiliation(s)
- Louise M Hesketh
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Catherine D E Wilder
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Niraja N Ranadive
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Georgia Lytra
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Patrisia Qazimi
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Jade S Munro
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Nakita Ahdi
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Michael J Curtis
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
14
|
Ueno N. The Effect of Sodium Channel Blocker, Mexiletine, on Body Weight in Type 2 Diabetes Patients with Visceral Obesity. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2019; 12:1179551418825049. [PMID: 30733634 PMCID: PMC6343451 DOI: 10.1177/1179551418825049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 12/05/2018] [Indexed: 12/28/2022]
Abstract
Objective: Mexiletine is an anti-arrhythmic agent also used for the treatment of painful diabetic neuropathy. In this study, the effect of mexiletine on body weight was evaluated in type 2 diabetes patients with diabetic neuropathy exhibiting visceral obesity. Methods: Type 2 diabetes patients with neuropathy exhibiting visceral obesity (n = 21) treated by mexiletine (300 mg/day) and a control group of type 2 diabetes patients with the same condition who received vitamin B12 (n = 12) were retrospectively evaluated. Body weight, waist circumference, hemoglobin A1c (HbA1c), blood pressure, liver function, serum lipids, and serum uric acid were assessed before and 6 months after the treatment. Results: Mexiletine significantly decreased body weight and waist circumference. The changes in body weight and waist circumference in 6 months in the mexiletine group were greater than in the control group. In metabolic parameters, there were significant decreases in triglyceride (TG) and serum uric acid. There were positive relationships between the change in body weight and the changes in TG, uric acid, alanine aminotransferase (ALT), and HbA1c. Conclusions: Mexiletine may affect body weight regulation. It ameliorated the metabolic parameters possibly by decreasing visceral fat. Further study should be performed to clarify the mechanism of the effect.
Collapse
Affiliation(s)
- Naohiko Ueno
- Ueno Internal Medicine and Diabetes Clinic, Kobe, Japan
| |
Collapse
|
15
|
Interactions of Mexiletine with Novel Antiepileptic Drugs in the Maximal Electroshock Test in Mice: An Isobolographic Analysis. Neurochem Res 2018; 43:1887-1896. [PMID: 30117096 PMCID: PMC6182375 DOI: 10.1007/s11064-018-2606-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/05/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Abstract
The aim of the study was to evaluate precisely the type of interactions between mexiletine (an antiarrhythmic drug) and four new generation antiepileptic drugs: lamotrigine, oxcarbazepine, topiramate and pregabalin in the maximal electroshock test in mice (MES). The isobolographic analysis was used to assess the nature of interactions between the tested drugs. Total brain concentrations of antiepileptics were also measured to detect possible pharmacokinetic interactions. The results obtained indicated that the mixture of mexiletine and pregabalin at the fixed ratios of 1:1 and 3:1 led to supra-additive interaction in terms of seizure suppression, while the proportion of 1:3 occurred additive. Synergism was also demonstrated for the combination of mexiletine and topiramate in all three proportions. Combinations of mexiletine with lamotrigine and mexiletine with oxcarbazepine were found to be additive. Adverse-effect profiles of mexiletine, antiepileptics and drug combinations were evaluated in the chimney test (motor coordination) and step-through passive-avoidance task (long-term memory). Mexiletine and drug combinations did not impair long-term memory. Moreover, all combinations of mexiletine with lamotrigine, oxcarbazepine and topiramate had no significant effect on motor coordination. However, the results from the chimney test indicated that pregabalin, administered alone at its ED50 dose from the MES-test, significantly impaired motor performance. Similar adverse effects were observed when mexiletine was co-administered with pregabalin at the fixed-dose ratio combinations of 1:1 and 1:3. However, reduction of pregabalin dose at the fixed ratio of 3:1 seems to prevent significant motor impairment. The results may indicate that mexiletine can be considered as an adjunctive drug in antiepileptic treatment, particularly in patients with concomitant cardiac arrhythmia.
Collapse
|
16
|
Life Threatening Severe QTc Prolongation in Patient with Systemic Lupus Erythematosus due to Hydroxychloroquine. Case Rep Cardiol 2016; 2016:4626279. [PMID: 27478650 PMCID: PMC4960328 DOI: 10.1155/2016/4626279] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/15/2016] [Indexed: 11/24/2022] Open
Abstract
We present a case of a syncopal episode resulting from significant QT interval prolongation in a patient on hydroxychloroquine for the treatment of systemic lupus erythematosus and end stage renal disease. The patient had been treated with hydroxychloroquine for two years prior to presentation. After thorough workup for secondary causes of QT interval prolongation hydroxychloroquine was discontinued and the patient's QT interval shortened. The patient was treated with mexiletine to prevent sudden ventricular arrhythmias, which was unique compared to other documented cases in which lidocaine was used. The patient was noted to have mild prolongation of the QT interval on electrocardiogram prior to initiation of hydroxychloroquine therapy which was exacerbated by its use and may have been caused due to toxicity from underlying renal failure.
Collapse
|
17
|
Borowicz-Reutt KK, Banach M, Piskorska B. Mexiletine and its Interactions with Classical Antiepileptic Drugs: An Isobolographic Analysis. Neurochem Res 2016; 41:1185-91. [PMID: 26738990 DOI: 10.1007/s11064-015-1812-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/22/2015] [Accepted: 12/18/2015] [Indexed: 11/28/2022]
Abstract
Using the mouse maximal electroshock test, the reference model of tonic-clonic seizures, the aim of the present study was to determine the type of interaction between mexiletine (a class IB antiarrhythmic drug) and classical antiepileptics: valproate, carbamazepine, phenytoin, and phenobarbital. Isobolographic analysis of obtained data indicated antagonistic interactions between mexiletine and valproate (for fixed ratio combinations of 1:1 and 3:1). Additivity was observed between mexiletine and valproate applied in proportion of 1:3 as well as between mexiletine and remaining antiepileptics for the fixed ratios of 1:3, 1:1, and 3:1. Neither motor performance nor long-term memory were impaired by mexiletine or antiepileptic drugs regardless of whether they were administered singly or in combination. Mexiletine did not significantly affected brain concentrations of carbamazepine, phenobarbital or phenytoin. In contrast, the antiarrhythmic drug decreased by 23 % the brain level of valproate. This could be, at least partially, the reason of antagonistic interaction between the two drugs. In conclusion, the observed additivity suggests that mexiletine can be safely applied in epileptic patients treated with carbamazepine, phenytoin or phenobarbital. Because of undesirable pharmacodynamics and pharmacokinetic interactions with valproate, mexiletine should not be used in such combinations.
Collapse
Affiliation(s)
- Kinga K Borowicz-Reutt
- Independent Unit of Experimental Neuropathophysiology, Department of Pathophysiology, Medical University, Lublin, Poland.
| | - Monika Banach
- Independent Unit of Experimental Neuropathophysiology, Department of Pathophysiology, Medical University, Lublin, Poland
| | - Barbara Piskorska
- Independent Unit of Experimental Neuropathophysiology, Department of Pathophysiology, Medical University, Lublin, Poland
| |
Collapse
|
18
|
Romano G, Raffa GM, Ruggieri A, Sgarito G, Falletta C, Sciacca S, Pilato M, Clemenza F. Recurrent ventricular tachycardia in a patient with continuous flow left ventricle assist device: Successful management with radiofrequency ablation and medical treatment. Int J Cardiol 2015; 190:198-200. [DOI: 10.1016/j.ijcard.2015.04.141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 10/23/2022]
|
19
|
Bhushan R, Vashistha VK. Synthesis of variants of Marfey's reagent having d-amino acids as chiral auxiliaries and liquid-chromatographic enantioseparation of (RS)-Mexiletine in spiked plasma: Assessment and comparison with l-amino acid analogs. J Chromatogr A 2015; 1379:43-50. [DOI: 10.1016/j.chroma.2014.12.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 11/26/2022]
|
20
|
Cregg R, Cox JJ, Bennett DLH, Wood JN, Werdehausen R. Mexiletine as a treatment for primary erythromelalgia: normalization of biophysical properties of mutant L858F NaV 1.7 sodium channels. Br J Pharmacol 2014; 171:4455-63. [PMID: 24866741 PMCID: PMC4209151 DOI: 10.1111/bph.12788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/10/2014] [Accepted: 05/17/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The non-selective sodium channel inhibitor mexiletine has been found to be effective in several animal models of chronic pain and has become popular in the clinical setting as an orally available alternative to lidocaine. It remains unclear why patients with monogenic pain disorders secondary to gain-of-function SCN9a mutations benefit from a low systemic concentration of mexiletine, which does not usually induce adverse neurological side effects. The aim of this study was, therefore, to investigate the biophysical effects of mexiletine on the L858F primary erythromelalgia NaV 1.7 mutation in vitro. EXPERIMENTAL APPROACH Human wild-type and L858F-mutated NaV 1.7 channels were expressed in HEK293A cells. Whole-cell currents were recorded by voltage-clamp techniques to characterize the effect of mexiletine on channel gating properties. KEY RESULTS While the concentration-dependent tonic block of peak currents by mexiletine was similar in wild-type and L858F channels, phasic block was more pronounced in cells transfected with the L858F mutation. Moreover, mexiletine substantially shifted the pathologically-hyperpolarized voltage-dependence of steady-state activation in L858F-mutated channels towards wild-type values and the voltage-dependence of steady-state fast inactivation was shifted to more hyperpolarized potentials, leading to an overall reduction in window currents. CONCLUSION AND IMPLICATIONS Mexiletine has a normalizing effect on the pathological gating properties of the L858F gain-of-function mutation in NaV 1.7, which, in part, might explain the beneficial effects of systemic treatment with mexiletine in patients with gain-of-function sodium channel disorders.
Collapse
Affiliation(s)
- R Cregg
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK; UCL Centre for Anaesthesia, Critical Care and Pain Medicine, London, UK
| | | | | | | | | |
Collapse
|
21
|
Grewal AK, Jaggi AS, Rana AC, Singh N. Effect of neurosteroid modulation on global ischaemia-reperfusion-induced cerebral injury in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:485-91. [PMID: 24381496 PMCID: PMC3874434 DOI: 10.4196/kjpp.2013.17.6.485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 09/25/2013] [Accepted: 10/17/2013] [Indexed: 11/17/2022]
Abstract
The present study was designed to investigate the putative effect of neurosteroid modulation on global ischaemia-reperfusion-induced cerebral injury in mice. Bilateral carotid artery occlusion followed by reperfusion, produced a significant rise in cerebral infarct size along with impairment of grip strength and motor coordination in Swiss albino mice. Administration of carbamazepine (16 mg/kg, i.p.) before global cerebral ischaemia significantly attenuated cerebral infarct size and improved the motor performance. However, administration of indomethacin (100 mg/kg, i.p.) attenuated the neuroprotective effect of carbamazepine. Mexiletine (50 mg/kg, i.p.) did not produce significant neuroprotective effect. It may be concluded that the neuroprotective effect of carbamazepine may be due to increase in synthesis of neurosteroids perhaps by activating enzyme (3α HSD) as indomethacin attenuated the neuroprotective effect of carbamazepine. The sodium channel blocking effect of carbamazepine may not be involved in neuroprotection as mexiletine, a sodium channel blocker, did not produce significant neuroprotective effect.
Collapse
Affiliation(s)
- Amarjot Kaur Grewal
- Rayat and Bahra Institute of Pharmacy, Sahauran, Mohali-140104, Punjab, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, Punjab, India
| | - Avtar Chand Rana
- Rayat and Bahra Institute of Pharmacy, Sahauran, Mohali-140104, Punjab, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, Punjab, India
| |
Collapse
|
22
|
Shailesh F, Singla S, Sureddi R, Deshmukh AJ, Paydak H. A patient with mexiletine-related psychosis. Int J Gen Med 2011; 4:765-6. [PMID: 22114522 PMCID: PMC3219764 DOI: 10.2147/ijgm.s25381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Mexiletine is a commonly used Class IB (Vaughan William classification) antiarrhythmic drug. We report a case of mexiletine-induced psychosis that was successfully managed by decreasing the dose and using alternative medications for management of ventricular tachycardia.
Collapse
Affiliation(s)
- Fnu Shailesh
- Division of Cardiovascular Medicine, University of Arkansas for Medical Science, Little Rock, AR, USA
| | | | | | | | | |
Collapse
|
23
|
Gelzer AR, Kraus MS, Rishniw M, Hemsley SA, Moïse NS. Combination therapy with mexiletine and sotalol suppresses inherited ventricular arrhythmias in German shepherd dogs better than mexiletine or sotalol monotherapy: A randomized cross-over study. J Vet Cardiol 2010; 12:93-106. [DOI: 10.1016/j.jvc.2010.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 05/19/2010] [Accepted: 06/15/2010] [Indexed: 10/19/2022]
|
24
|
Affiliation(s)
- Shu-Feng Zhou
- Discipline of Chinese Medicine, School of Health Sciences, RMIT University, Victoria, Australia.
| |
Collapse
|
25
|
Tzanavaras PD, Zacharis CK, Rigas P. Novel automated assay for the quality control of mexiletine hydrochloride formulations using sequential injection and on-line dilution. J Pharm Biomed Anal 2008; 48:1254-60. [DOI: 10.1016/j.jpba.2008.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2008] [Revised: 09/02/2008] [Accepted: 09/04/2008] [Indexed: 10/21/2022]
|
26
|
Ulu ST. LC Method with Precolumn Derivatization for Analysis of Mexiletine in Pharmaceutical Preparations. Chromatographia 2008. [DOI: 10.1365/s10337-008-0897-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
El Yaman M, Perry J, Makielski JC, Ackerman MJ. Suppression of atrial fibrillation with mexiletine pharmacotherapy in a young woman with type 1 long QT syndrome. Heart Rhythm 2008; 5:472-4. [DOI: 10.1016/j.hrthm.2007.10.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Accepted: 10/13/2007] [Indexed: 11/28/2022]
|
28
|
Salinas FV, Malik K, Benzon HT. Local Anesthetics for Regional Anesthesia and Pain Management. RAJ'S PRACTICAL MANAGEMENT OF PAIN 2008:811-838. [DOI: 10.1016/b978-032304184-3.50047-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
29
|
Frederickson M, Callaghan O, Chessari G, Congreve M, Cowan SR, Matthews JE, McMenamin R, Smith DM, Vinković M, Wallis NG. Fragment-Based Discovery of Mexiletine Derivatives as Orally Bioavailable Inhibitors of Urokinase-Type Plasminogen Activator. J Med Chem 2007; 51:183-6. [DOI: 10.1021/jm701359z] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Martyn Frederickson
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Owen Callaghan
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Gianni Chessari
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Miles Congreve
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Suzanna R. Cowan
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Julia E. Matthews
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Rachel McMenamin
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Donna-Michelle Smith
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Mladen Vinković
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Nicola G. Wallis
- Astex Therapeutics Ltd, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| |
Collapse
|
30
|
Atalay B, Caner H, Can A, Cekinmez M. Attenuation of microtubule associated protein-2 degradation after mild head injury by mexiletine and calpain-2 inhibitor. Br J Neurosurg 2007; 21:281-7. [PMID: 17612919 DOI: 10.1080/02688690701364781] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The objective of the study was to address the early effects of mild, closed, head injuries on neuronal stability and the prevention of microtubule-associated protein-2 (MAP-2) degradation by mexiletine and calpain-2 inhibitor. Twenty-four rats were divided into four groups: control group (1); trauma group without treatment (2); mexiletine-pretreated and subjected to trauma group (3); trauma subjected and then calpain-2 inhibitor received group (4). All animals were subjected to mild, closed, head trauma. Frontal lobes were removed and processed for staining and immunofluorescent labelling of MAP-2 cytoskeletal proteins, which were evaluated by confocal microscopy in serial optical sections showing the three dimensional cytoarchitecture of affected areas. MAP-2 decoration in almost all neurons obtained from traumatized brain regions drastically diminished, while minute filamentous and granular profiles in axons and/or dendrites were retained together implying a massive degradation/depolymerization of microtubules. In contrast, in mexiletine-pretreated animals, MAP-2 positivity in axonal and perikaryonal profiles was fairly retained, which clearly depicts the protective role of mexiletine after trauma. Compared with mexiletine-pretreated group, calpain-2 inhibitor treated group displayed a less well-preserved MAP-2 expression. Mexiletine can prevent cytoskeletal structure and protein degradation after mild head trauma. Calpain-2 inhibitor prevents protein degradation, but cytoskeletal organization is better preserved with mexiletine.
Collapse
Affiliation(s)
- B Atalay
- Department of Neurosurgery, Baskent University Faculty of Medicine, Ankara, Turkey.
| | | | | | | |
Collapse
|
31
|
Sen O, Caner H, Aydin MV, Ozen O, Atalay B, Altinors N, Bavbek M. The effect of mexiletine on the level of lipid peroxidation and apoptosis of endothelium following experimental subarachnoid hemorrhage. Neurol Res 2007; 28:859-63. [PMID: 17288746 DOI: 10.1179/016164106x115099] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE The role of apoptosis in etiopathogenesis of vasospasm is not clearly understood yet. It is widely accepted that protection of the endothelial cells from the process of apoptosis could have beneficial effects on cerebral vasospasm after subarachnoid hemorrhage (SAH). Mexiletine blocks sodium and calcium channels and activates ATP-sensitive K(+) channels. Moreover, mexiletine is known to have potent antioxidant effects through inhibiting free-radical production. METHODS Twenty-one rabbits were allocated into three groups randomly. Group I was sham operated group (n=7). SAH occurred but no medication was given to the Group II rabbits (SAH only group) (n=7). Mexiletine (50 mg/kg, b.i.d., i.p.) was administered just before SAH and continued until 48 hours following SAH to the Group III rabbits (Mexiletine treated group) (n=7). The ApopTag peroxidase in situ apoptosis detection kit (Serologicals Corporation, former Intergen) was used to demonstrate apoptosis in a cross section of basillary arteries. Thiobarbituric acid reactive material was used to determine the lipid peroxidation levels. RESULTS There was a statistically significant difference between lipid peroxidation product levels of the control and SAH only groups (p<0.05). The level of lipid peroxidation production in Mexiletine treated group was significantly lower compared with SAH only group (p<0.05) but not significantly higher than the control group (p>0.05). DISCUSSION In the present study we investigated the antioxidant action of mexiletine on apoptosis of endothelium following a rabbit SAH model. This experimental study directly suggested that lipid peroxidation is an important step in development of apoptosis in endothelial cells and prevention of structural integrity of endothelial cell should play a beneficial role in attenuation of cerebral vasospasm. Mexiletine treatment prevented the increase in lipid peroxidation and cerebral vasospasm. Examination of endothelial cells by staining specific for apoptosis demonstrated significant protection of cell integrity in the treated group.
Collapse
Affiliation(s)
- Orhan Sen
- Department of Neurosurgery, Baskent University, Ankara, Turkey.
| | | | | | | | | | | | | |
Collapse
|
32
|
Kunicki PK, Sitkiewicz D. High Performance Liquid Chromatographic Analysis of Some Antiarrhythmic Drugs in Human Serum Using Cyanopropyl Derivatized Silica Phase. J LIQ CHROMATOGR R T 2006. [DOI: 10.1080/10826079608006310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Paweł K. Kunicki
- a Department of Clinical Biochemistry , National Institute of Cardiology , Alpejska 42 04-628, Warszawa , Poland
| | - Dariusz Sitkiewicz
- a Department of Clinical Biochemistry , National Institute of Cardiology , Alpejska 42 04-628, Warszawa , Poland
| |
Collapse
|
33
|
Takada M, Goto T, Kotake T, Saito M, Kawato N, Nakai M, Gunji T, Shibakawa M. Appropriate dosing of antiarrhythmic drugs in Japan requires therapeutic drug monitoring*. J Clin Pharm Ther 2005; 30:5-12. [PMID: 15658999 DOI: 10.1111/j.1365-2710.2004.00612.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE In general, drugs are used in accordance with an approved dosage regimen in expectation of an appropriate balance between efficacy and toxicity. However, dose control of drugs with a narrow therapeutic range and marked intersubject variability in pharmacokinetics should be established through individualization of dosing based on therapeutic drug monitoring (TDM). The purpose of this study was to examine differences between the approved dosage regimen and the doses of antiarrhythmic drugs and digoxin used in clinical practice and to examine the influence of TDM on dosing. METHODS Prescription research of antiarrhythmic drugs was performed at five national hospitals in Japan. Prescriptions for antiarrhythmic drugs (cibenzoline, disopyramide, pirmenol, mexiletine, aprindine, flecainide, pilsicainide, amiodarone and digoxin) were counted for the study period. The mean dose and dose distribution of the drugs were determined in each hospital. Comparisons were made of mean dose obtained in the study with the dosage approved by the authority. In addition, the percentage of patients that received TDM was determined. RESULTS A difference was seen between the approved dosage and the actual dose. For all drugs except flecainide, the mean dose was smaller than the approved dosage. For all drugs except digoxin, remarkable variations were seen in the dose distribution among the hospitals. Digoxin showed a similar dose distribution among the five hospitals. Overall, the percentage of patients that received TDM was low except for Hospital A. However, TDM of digoxin was relatively common at four of the hospitals. CONCLUSIONS It is concluded that, with the exception of digoxin, the appropriate dosing regimen for antiarrhythmic drugs is not yet established. The establishment of appropriate dosing regimens for antiarrhythmic drugs requires the more widespread adoption of TDM.
Collapse
Affiliation(s)
- M Takada
- Department of Pharmacy, National Cardiovascular Center, Suita-city, Osaka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Kohjitani A, Miyawaki T, Funahashi M, Mitoh Y, Matsuo R, Shimada M. Mexiletine inhibits nonadrenergic noncholinergic lower oesophageal sphincter relaxation in rabbits. Eur J Pharmacol 2003; 465:145-51. [PMID: 12650844 DOI: 10.1016/s0014-2999(03)01483-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nonadrenergic noncholinergic (NANC) nerves are known to be nitrergic and to have an important role in the regulation of gastrointestinal motility and function. Cardiac antiarrhythmic therapy in humans is accompanied by a high incidence of gastrointestinal side-effects. We investigated the effect of mexiletine, a class Ib antiarrhythmic drug, on NANC lower oesophageal sphincter relaxation. Mexiletine concentration dependently inhibited the NANC relaxation induced by 30 mM KCl (EC(50)=4.4 x 10(-6) M); the production of 3',5'-cyclic guanosine monophosphate (cGMP) after KCl stimulation was concentration dependently decreased. The relaxation induced by the exogenous nitric oxide (NO) donor, diethylamine NONOate (10(-5) M), was not inhibited by mexiletine, and the cGMP production after diethylamine NONOate application was not altered. Mexiletine did not alter the activity of NO synthase. These findings suggest that mexiletine inhibits NANC relaxation via NO-cGMP pathway modulation, possibly by inhibiting myenteric nitrergic neurotransmission in the lower oesophageal sphincter in rabbits.
Collapse
Affiliation(s)
- Atsushi Kohjitani
- Department of Dental Anaesthesiology, Okayama University Graduate School of Medicine and Dentistry, 2-5-1 Shikata-cho, Okayama 700-8525, Japan.
| | | | | | | | | | | |
Collapse
|
36
|
Takahashi MP, Cannon SC. Mexiletine block of disease-associated mutations in S6 segments of the human skeletal muscle Na(+) channel. J Physiol 2001; 537:701-14. [PMID: 11744749 PMCID: PMC2278988 DOI: 10.1111/j.1469-7793.2001.00701.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
1. Over twenty different missense mutations in the alpha-subunit of the adult skeletal muscle Na(+) channel (hSkM1) have been identified as a cause of myotonia or periodic paralysis. We examined state-dependent mexiletine block for mutations involving the putative binding site in S6 segments (V445M, S804F, V1293I, V1589M and M1592V). Whole-cell Na(+) currents were measured from wild-type (WT) and mutant channels transiently expressed in HEK cells. 2. Use-dependent block (10 ms pulses to -10 mV, at 20 Hz) in 100 microM mexiletine was reduced modestly by mutations in IVS6 (V1589M, M1592V) and enhanced by the mutation in IS6 (V445M). For mutations in IIS6 (S804F) and IIIS6 (V1293I) use-dependent block was not statistically different from that of wild-type channels. 3. Resting-state block (10 ms pulses to -10 mV from -150 mV, at 0.1 Hz) of S6 mutants was comparable to that of WT (dissociation constant for resting channels, K(R) = 650 +/- 40 microM, n = 9). The S6 mutant with the greatest change in K(R) was V445M (K(R) = 794 +/- 45 microM, n = 5), but this difference was only marginally significant (P = 0.047). 4. A modified technique for estimating local anaesthetic affinity of inactivated channels was developed to reduce errors due to slow inactivation and to failure of drug binding to reach equilibrium. Mexiletine affinity for inactivated channels was reduced by mutations in IVS6 (V1589M: dissociation constant for the inactivated state (K(I)) = 44.7 microM; M1592V: K(I) = 40.0 microM) and increased by the mutation in IS6 (V445M: K(I) = 15.0 microM), compared to wild-type channels (K(I) = 28.3 microM). 5. We conclude that the disease-associated S6 mutations in domains I-IV cause at most a 2-fold change in inactivated state affinity and have even less of an effect on resting block. Model simulations show that the reduced use-dependent block of IVS6 mutants derives primarily from an increased off-rate at hyperpolarized potentials, whereas the enhanced use-dependent block of the IS6 mutant was due to a higher affinity for inactivated V445M channels.
Collapse
Affiliation(s)
- M P Takahashi
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
37
|
Lanchote VL, Bonato PS, Cesarino EJ, Mere Júnior YA, Cavani SR, Santos J, Bertucci C. Enantioselective determination of the hydroxylated metabolites of mexiletine in human plasma. Chirality 2000; 9:732-8. [PMID: 9435098 DOI: 10.1002/(sici)1520-636x(1997)9:8<732::aid-chir4>3.0.co;2-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pre-column derivatization with o-phthaldialdehyde and N-acetyl-l-cysteine was used for liquid-chromatographic diastereomeric resolution of p-hydroxymexiletine (PHM) and hydroxymethylmexiletine (HMM), metabolites of mexiletine formed by aromatic and aliphatic hydroxylation, respectively. The resulting diastereomeric derivatives were resolved on a C18 column and monitored by fluorescence detection. The diastereomeric elution order for both metabolites was determined on the basis of the circular dichroism spectra of each eluted fraction. Plasma samples (500 microliters) showed recoveries greater than 75% for both the metabolites. Calibration curves in plasma samples were linear over the concentration ranges 10-500 and 20-1,000 ng/ml for each enantiomer of PHM and HMM, respectively. The limits of quantitation were found to be 10.0 and 5.0 ng/ml for both enantiomers of PHM and HMM. The within-day and between-day coefficients of variation were less than 10%. The assay was shown to be suitable for a pharmacokinetic study performed in a patient with ventricular arrhythmias following the short-term oral treatment of 200 mg t.i.d. of racemic mexiletine hydrochloride.
Collapse
Affiliation(s)
- V L Lanchote
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
38
|
Saitoh M, Aye NN, Komori S, Nakazawa T, Hashimoto K. Effects of HNS-32, a novel antiarrhythmic drug, on ventricular arrhythmias induced by coronary artery occlusion and reperfusion in anesthetized rats. Mol Cell Biochem 2000; 205:133-40. [PMID: 10821431 DOI: 10.1023/a:1007074115503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
HNS-32 (N1,N1-dimethyl-N2-(2-pyridylmethyl)-5-isopropyl-3, 8-dimethylazulene-1-carboxamidine: CAS 186086-10-2) is a newly synthesized compound, and possesses antiarrhythmic properties with vasodilator action in dog hearts. The aim of this study was to investigate the dose-dependent effects of HNS-32 on ischemia- and/or reperfusion-induced ventricular arrhythmias in anesthetized rats in vivo and compared with those of mexiletine. Saline or drugs were administered intravenously 5 min prior to coronary artery occlusion. On the ischemia-induced ventricular arrhythmias, HNS-32 showed dose-dependent reduction of total number of premature ventricular complexes (PVC) from 2091+/-225 to 656+/-116 and 286+/-69 beats/30 min (p < 0.05), the ventricular tachycardia (VT) duration from 183+/-33 to 28+/-9 and 4+/-2 sec (p < 0.05), the incidence of VT from 100 to 90 (n.s.) and 40% (p < 0.05), and the incidence of ventricular fibrillation (VF) from 50 to 0 and 0% (p < 0.05) with 3 and 5 mg/kg, respectively. Mexiletine also reduced these parameters to 936+/-159 beats/30 min (p < 0.05), 39+/-22 sec (p < 0.05), 90% (n.s.) and 10% (n.s.), respectively. HNS-32 completely suppressed the late reperfusion-induced arrhythmias, however mexiletine did not affect them. On the early reperfusion-induced ventricular arrhythmias, HNS-32 showed dose-dependent reduction of VT duration from 126+/-34 to 37+/-12 and 3+/-2 sec (p < 0.05), incidence of VT from 100 to 90 (n.s.) and 40% (p < 0.05), incidence of VF from 100 to 10 and 0% (p < 0.05), and mortality rate from 90 to 0 and 0% (p < 0.05), with 3 and 5 mg/kg, respectively. Mexiletine also reduced these parameters to 16+/-9 sec (p < 0.05), 80 (n.s.), 50 (p < 0.05), and 10% (p < 0.05), respectively. HNS-32 significantly reduced the heart rate in a dose-dependent manner, from 399+/-14 to 350+/-8 and 299+/-10 beats/min (p < 0.05) with 3 and 5 mg/kg, respectively. The antiarrhythmic effects of HNS-32 were more potent than that of the similar dose of mexiletine against occlusion-induced and reperfusion-induced arrhythmias in in vivo rats.
Collapse
Affiliation(s)
- M Saitoh
- Department of Pharmacology, Yamanashi Medical University, Japan
| | | | | | | | | |
Collapse
|
39
|
Abstract
Liver disease can modify the kinetics of drugs biotransformed by the liver. This review updates recent developments in this field, with particular emphasis on cytochrome P450 (CYP). CYP is a rapidly expanding area in clinical pharmacology. The information currently available on specific isoforms involved in drug metabolism has increased tremendously over the latest years, but knowledge remains incomplete. Studies on the effects of liver disease on specific isoenzymes of CYP have shown that some isoforms are more susceptible than others to liver disease. A detailed knowledge of the particular isoenzyme involved in the metabolism of a drug and the impact of liver disease on that enzyme can provide a rational basis for dosage adjustment in patients with hepatic impairment. The capacity of the liver to metabolise drugs depends on hepatic blood flow and liver enzyme activity, both of which can be affected by liver disease. In addition, liver failure can influence the binding of a drug to plasma proteins. These changes can occur alone or in combination; when they coexist their effect on drug kinetics is synergistic, not simply additive. The kinetics of drugs with a low hepatic extraction are sensitive to hepatic failure rather than to liver blood flow changes, but drugs having a significant first-pass effect are sensitive to alterations in hepatic blood flow. The drugs examined in this review are: cardiovascular agents (angiotensin converting enzyme inhibitors, angiotensin II receptor antagonists, calcium antagonists, ketanserin, antiarrhythmics and hypolipidaemics), diuretics (torasemide), psychoactive and anticonvulsant agents (benzodiazepines, flumazenil, antidepressants and tiagabine), antiemetics (metoclopramide and serotonin antagonists), antiulcers (acid pump inhibitors), anti-infectives and antiretroviral agents (grepafloxacin, ornidazole, pefloxacin, stavudine and zidovudine), immunosuppressants (cyclosporin and tacrolimus), naltrexone, tolcapone and toremifene. According to the available data, the kinetics of many drugs are altered by liver disease to an extent that requires dosage adjustment; the problem is to quantify the required changes. Obviously, this requires the evaluation of the degree of hepatic impairment. At present there is no satisfactory test that gives a quantitative measure of liver function and its impairment. A critical evaluation of these methods is provided. Guidelines providing a rational basis for dosage adjustment are illustrated. Finally, it is important to consider that liver disease not only affects pharmacokinetics but also pharmacodynamics. This review also examines drugs with altered pharmacodynamics.
Collapse
Affiliation(s)
- V Rodighiero
- Department of Pharmacology, University of Padova, Italy
| |
Collapse
|
40
|
Kaptanoglu E, Caner HH, Sürücü HS, Akbiyik F. Effect of mexiletine on lipid peroxidation and early ultrastructural findings in experimental spinal cord injury. J Neurosurg 1999; 91:200-4. [PMID: 10505505 DOI: 10.3171/spi.1999.91.2.0200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The purpose of this study was to investigate the effect of mexiletine on lipid peroxidation and on ultrastructural findings after induced spinal cord injury (SCI). The authors also compared the activity of mexiletine to that of the well-known antioxidant, methylprednisolone sodium succinate (MPSS). METHODS Wistar rats were divided into seven groups, (Groups 1-7). Those in Groups 1 and 2 were control animals that underwent laminectomy only, after which nontraumatized spinal cord samples were obtained immediately (Group 1) and 2 hours postsurgery (Group 2). Spinal cord injury was induced in all other groups, and cord samples were obtained at 2 hours postsurgery. The rats in Group 3 underwent SCI alone; those in Group 4 received 30 mg/kg of MPSS intraperitoneally immediately after trauma was induced; and those in Groups 5, 6, and 7 received 1, 10, and 50 mg/kg of mexiletine, respectively, by intraperitoneal injection immediately after trauma was induced. Compared with the levels in control animals, lipid peroxidation was significantly elevated in rats in Groups 3 and 5, but there were no statistical differences among those in Groups 1, 2, 4, 6 and 7 in this regard. Compared with the findings in rats in Group 3, ultrastructural damage post-SCI was minor in rats in Groups 4 and 5, and there was even less damage evident in rats in Group 7. CONCLUSIONS Analysis of these findings showed that administration of 50 mg/kg mexiletine significantly decreased the level of lipid peroxidation and protected spinal cord ultrastructure following SCI.
Collapse
Affiliation(s)
- E Kaptanoglu
- Department of Neurosurgery, Ankara Numune Hospital, Turkey
| | | | | | | |
Collapse
|
41
|
Abstract
UNLABELLED Mexiletine is an orally active local anaesthetic agent which is structurally related to lidocaine (lignocaine) and has been used for alleviating neuropathic pain of various origins. Mexiletine has been evaluated in several randomised, placebo-controlled trials in patients with painful diabetic neuropathy. The drug decreased mean visual analogue scale (VAS) pain ratings in all studies that used this measure, although in only 2 studies was this effect significantly greater than the often substantial responses seen with placebo. The clinical significance of these decreases is not clear. Statistically significant (vs placebo) reductions in VAS pain ratings were observed in 16 patients receiving mexiletine 10 mg/kg/day for 10 weeks in 1 study and in nocturnal (but not diurnal) pain in 31 patients receiving mexiletine 675 mg/day for 3 weeks in another. Retrospective analysis of another study revealed that mexiletine recipients (225 to 675 mg/day) who described their pain as stabbing, burning or formication on the pain-rating-index-total instrument of the McGill Pain Questionnaire, experienced statistically significant reductions in VAS pain scores after 5 weeks, compared with placebo recipients. Mexiletine generally did not have a significant influence on the quality of sleep in patients with diabetic neuropathy. In Japanese patients, statistically significant reductions in subjective pain ratings were achieved with mexiletine 300 mg/day in 1 study and with 450 mg/day in a further study. In controlled trials, the frequency of adverse events in patients receiving mexiletine for painful diabetic neuropathy ranged from 13.5 to 50%. Gastrointestinal complaints, of which nausea was the most frequent, were the most common adverse events in mexiletine recipients. Central nervous system complaints were uncommon, but included: sleep disturbance, headache, shakiness, dizziness and tiredness. Serious cardiac arrhythmias have not been reported in patients receiving mexiletine for painful diabetic neuropathy; however, transient tachycardia and palpitations have been reported. There are significant differences in the metabolism of mexiletine between people who have cytochrome P450 2D6 [CYP2D6; extensive metabolisers (EMs)] and those who lack this isoenzyme [poor metabolisers (PMs)]. EMs, but not PMs, are susceptible to drug interactions between mexiletine and drugs that inhibit CYP2D6 (e.g. quinidine). Moreover, mexiletine inhibits CYP2D6-mediated metabolism of metoprolol and cytochrome P450 1A2-mediated metabolism of theophylline. Phenytoin and rifampicin (rifampin) induce the metabolism of mexiletine. Clearance of mexiletine is impaired in patients with hepatic, but not renal, dysfunction. Hence, dosage adjustments may be necessary in patients with liver disease. CONCLUSIONS Tricyclic antidepressants (TCAs) are the agents of choice for painful diabetic neuropathy; however, they are ineffective in approximately 50% of patients and are generally not well tolerated. Mexiletine is an alternative agent for the treatment of painful diabetic neuropathy in patients who have not had a satisfactory response to, or cannot tolerate, TCAs and/or other drugs.
Collapse
Affiliation(s)
- B Jarvis
- Adis International Limited, Auckland, New Zealand.
| | | |
Collapse
|
42
|
Lanchote VL, Cesarino EJ, Santos VJ, Moraes Júnior AV, Zanardi AM, Santos SR. Stereoselective metabolism of mexiletine in Chagasic women with ventricular arrhythmias. Eur J Drug Metab Pharmacokinet 1998; 23:259-66. [PMID: 9725491 DOI: 10.1007/bf03189349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Following a week of racemic mexiletine HCl at 200 mg tid (2x100 mg capsules), stereoselective aliphatic hydroxylation was studied in eight Chagasic women with chronic ventricular arrhythmias (52-67 yrs) with no history of renal or hepatic diseases. Blood samples were collected at dose interval up to 24 h of drug administration. Plasma concentrations of R(-) and S(+) mexiletine (MEX) and its metabolite hydroxymethylmexiletine (HMM) were determined by HPLC after derivatization with chiral reagent. The differences between R(-) and S(+) enantiomers were compared by paired t-test. Results are mean (95% CI). The following differences (p < 0.05) between R(-) and S(+) enantiomers, respectively, were found: MEX AUCss(0-8) 2.34 (1.84-2.85) vs 2.55 (1.97-3.13) microg.ml(-1) x h(-1); MEX CL/f 11.27 (7.77-14.77) vs 10.46 (7.18-13.74)ml.min(-1).Kg(-1); HMM Cmax 38.26 (24.3-52.22) vs 16.73 (10.1-23.29)ng.ml(-1); HMM Tmax 4.71 (2.67-6.76) vs 3.29 (1.24-5.33) h and HMM AUCss(0-8) 253.50 (165.39-341.61) vs 103.70 (69.51-137.90)ng.ml(-1).h(-1). The AUCss(0-8) ratio R(-)/S(+) for MEX was 0.93 (0.87-0.98) while for HMM was 2.50 (2.16-2.85). Distribution of MEX and HMM enantiomers were not significantly different. In this study we demonstrate that kinetic disposition of mexiletine exhibits stereoselectivity in vivo and that aliphatic hydroxylation is favored for R(-) mexiletine in Chagasic women with ventricular arrhythmias.
Collapse
Affiliation(s)
- V L Lanchote
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
43
|
Lanchote VL, Bonato PS, Dreossi SA, Gonçalves PV, Cesarino EJ, Bertucci C. High-performance liquid chromatographic determination of mexiletine enantiomers in plasma using direct and indirect enantioselective separations. JOURNAL OF CHROMATOGRAPHY. B, BIOMEDICAL APPLICATIONS 1996; 685:281-9. [PMID: 8953169 DOI: 10.1016/s0378-4347(96)00195-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Two methods were developed for the determination of mexiletine enantiomers in plasma samples suitable for studies on the stereoselective disposition of this drug. Both methods used fluorescence detection to improve sensitivity and selectivity. The direct enantioselective separation was based on the chiral resolution of mexiletine-2-naphthamide derivatives on a Chiralcel OJ column. The calibration curves were linear over the concentration range 50-500 ng/ml for each enantiomer; therefore the method can be used only for therapeutic monitoring, drug interaction and multiple dose pharmacokinetic studies. The indirect method was based on the formation of diastereomers using o-phthaldialdehyde and N-acetyl-L-cysteine reagents. The diastereomers were resolved on a reversed-phase RP-18 column. The method proved to be suitable for single or multiple dose pharmacokinetic studies based on the low quantification limit (1 ng/ml) and the broader linear range (1-1000 ng/ml) obtained.
Collapse
Affiliation(s)
- V L Lanchote
- Faculdade de Ciências Farmacêuticas de Ribeirão, Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | | | | | | | | |
Collapse
|
44
|
Kamiyama T, Tanonaka K, Harada H, Nakai K, Takeo S. Mexiletine and lidocaine reduce post-ischemic functional and biochemical dysfunction of perfused hearts. Eur J Pharmacol 1995; 272:151-8. [PMID: 7713159 DOI: 10.1016/0014-2999(94)00640-s] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The present study was undertaken to determine whether class Ib antiarrhythmic agents, mexiletine and lidocaine, exert beneficial effects on ischemia/reperfusion-induced cardiac contractile dysfunction. Isolated rat hearts were subjected to 35-min global ischemia, followed by 60-min reperfusion and the functional and metabolic alterations were examined with and without mexiletine or lidocaine treatment. Ischemia/reperfusion resulted in a lack of recovery of contractile function, a sustained rise in left ventricular end-diastolic pressure and increased coronary perfusion pressure of the perfused heart during reperfusion. Contractile dysfunction was associated with increases in tissue Na+ and Ca2+ levels, decreases in K+ and Mg2+ levels, and release of creatine kinase and purine nucleosides and bases (ATP metabolites) from the heart. Treatment of the perfused heart with either 10-100 microM of either mexiletine or lidocaine during pre-ischemia resulted in an enhancement of post-ischemic contractile recovery, a suppression of changes in tissue Na+, K+, Ca2+ and Mg2+ contents and an attenuation of the release of creatine kinase and ATP metabolites in an almost concentration-dependent manner. Tissue sodium accumulation was observed at the end of ischemia, which was also attenuated by pretreatment with these agents. The prevention of Na+ overload and accompanying Ca2+ overload in cardiac cells may be the mechanism underlying the improvement of post-ischemic contractile function of perfused hearts by these agents.
Collapse
Affiliation(s)
- T Kamiyama
- Department of Pharmacology, Tokyo College of Pharmacy, Japan
| | | | | | | | | |
Collapse
|
45
|
Abstract
Many of the antiarrhythmic drugs produce a rise in the ventricular defibrillation threshold (DFT). Although mexiletine has also been reported as the probable cause of a significant elevation of DFT, there has been no previous study; therefore, the effect of mexiletine on DFT was investigated in the present study. The experiments were performed on ten mongrel dogs in the open-chest state using general anesthesia. Mexiletine 1, 2, 4, 6, or 8 mg/kg was administered as the loading dose, followed by the same dose/kg per hour. In these five groups, fibrillation/defibrillation (F/D) trials were performed repeatedly every 10 minutes, until 60 minutes after starting the maintenance dose. F/D trials were also performed at 30, 45, and 60 minutes after the completion of mexiletine infusion. The heart was allowed to fibrillate for a total of 30 seconds. Applying internal paddles to the heart, energies of 2, 3, 5, 7, 10, 20, and 30 J maximum were used. The minimal energy shock that caused defibrillation was defined as the DFT. The mexiletine concentration in each group changed from 0 to 6.11 micrograms/mL, DFT ranged from 2-10 J, and no statistical correlation was found between mexiletine concentration and DFT. We conclude that mexiletine does not induce an increase in DFT in dogs.
Collapse
Affiliation(s)
- S Sato
- Department of Anesthesiology, University of Tsukuba, Ibaraki, Japan
| | | | | |
Collapse
|
46
|
Zaliunas R, Zabiela P, Slapikas R, Vainoras A, Pentiokiniene D, Levisauskiene R, Bechtold H, Meyer U. The effects of intravenous mexiletine on spectra of the signal-averaged ECG. Pacing Clin Electrophysiol 1994; 17:2187-93. [PMID: 7845841 DOI: 10.1111/j.1540-8159.1994.tb03824.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
This study examined the effects of intravenous mexiletine on the time-domain and spectrotemporal signal-averaged ECG (SAECG). SAECGs were recorded in 60 postmyocardial infarction (MI) patients with more than 100 premature ventricular beats per hour, before and after a constant infusion of mexiletine, 7 mg/kg, given over 1 hour. Spectrotemporal analysis was done on a fixed analyzed signal duration of QRS complex and ST segment of X, Y, and Z leads using a temporal window of rectangular type, measuring the signal content between 10-120 Hz. Intravenous mexiletine produced no significant change in the mean values of any of the time-domain variables. However, mexiletine either increased or decreased the power density spectrum (PDS) and PDS ratio (40-120 Hz/25-120 Hz) of the SAECG, and in rare cases only did it not alter the spectra of the SAECG. When PDS ratio in lead Z decreased after mexiletine, it was usually higher at baseline in comparison with that when the PDS ratio increased, or vice versa. When the treatment with mexiletine was effective, PDS increased in comparison with that when the drug was ineffective, or vice versa. Not all ranges (10-120 Hz) of spectra of the SAECG, but only certain frequency bands (25-40 Hz in lead Z, P = 0.0253; 40-55 Hz in lead Y, P = 0.0096; 55-70 Hz in X lead, P = 0.0018; and in lead Z, P = 0.0002; 70-85 Hz in lead X, P = 0.0025; and in lead Z, P = 0.0075, 85-100 Hz in lead X, P = 0.0033) were associated with mexiletine efficacy.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- R Zaliunas
- Department of Cardiology, Kaunas Medical Academy, Lithuania
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Zaliunas R, Zabiela P, Slapikas R, Vainoras A, Pentiokiniene D, Levisauskiene R, Bechtold H, Meyer U. Signal-averaged ECG in prediction of the short-term suppression of ventricular premature beats by Mexiletine. Int J Cardiol 1994; 46:243-54. [PMID: 7529214 DOI: 10.1016/0167-5273(94)90247-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We analyzed whether baseline parameters of time-domain and spectrotemporal analysis of a signal-averaged ECG or their changes during Mexiletine therapy can predict the antiarrhythmic efficacy of the drug. On 60 post-MI patients with > 100 ventricular premature beats per hour, signal-averaged ECGs were recorded before and after a constant infusion of Mexiletine (7 mg/kg) for 1 h and again after 4 days of oral Mexiletine therapy (Mexiletine SR, 360 mg twice daily). Spectrotemporal analysis was performed on a fixed analyzed signal duration of QRS-complex and ST-segment of X-, Y-, Z-leads using the temporal window of the rectangular type, measuring signals between 10-20 Hz. Intravenous and oral Mexiletine did not produce significant changes in mean values of any time-domain parameters. However, using informative variables of spectra of the signal-averaged ECG, we managed retrospectively to predict antiarrhythmic efficacy in 92% of the patients. Only certain frequency bands (from the range of the spectra at baseline, 10-120 Hz) were predictive for intravenous Mexiletine efficacy: 40-55 Hz in lead Y (P = 0.0116); 55-70 Hz in leads X and Z (P = 0.0063 and P = 0.0269, respectively); 70-85 Hz in lead Z, (P = 0.0227). When the treatment with intravenous Mexiletine was effective, the baseline power spectrum density was lower than when the drug was ineffective, and vice versa. Moreover, the efficacy of oral Mexiletine can be predicted by power density spectrum at baseline (10-25 Hz in lead Z, P = 0.0210; 70-85 Hz in lead Y, P = 0.0254) and by one of the possible (increased, decreased, unchanged) effects of intravenous Mexiletine on the spectra at frequency bands (70-85 Hz in lead X, P = 0.0432 and 40-120 Hz in lead Z, P = 0.0156). These results show the value of spectrotemporal signal-averaged ECG in selecting a subgroup of post-myocardial infarction patients that may benefit from Mexiletine therapy.
Collapse
Affiliation(s)
- R Zaliunas
- Department of Cardiology, Kaunas Medical Academy, Lithuania
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dohi Y, Kojima M, Sato K. Vasorelaxant effect of mexiletine in mesenteric resistance arteries of rats. Br J Pharmacol 1994; 111:673-80. [PMID: 8019745 PMCID: PMC1910102 DOI: 10.1111/j.1476-5381.1994.tb14790.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
1. The vascular action of mexiletine, a class Ib antiarrhythmic agent, was investigated in the mesenteric resistance arteries of rats. 2. The second order branch of the mesenteric artery was cut into rings and changes in isometric tension were recorded. 3. Mexiletine (10(-6) -10(-3) M) evoked concentration-dependent, endothelium-independent relaxations in arteries contracted with noradrenaline. 4. Mexiletine (10(-4) M) did not affect the contraction induced by noradrenaline in Ca(2+)-free solution, while the compound inhibited the contraction induced by CaCl2 in noradrenaline-activated arteries. 5. The relaxation induced by mexiletine was less pronounced in arteries contracted with high KCl than in those contracted with noradrenaline. 6. Mexiletine induced identical relaxations in arteries contracted with noradrenaline in high KCl solution containing verapamil and in Krebs solution. 7. Thus, mexiletine induces relaxations by inhibiting transmembrane Ca2+ movement, but not Ca2+ release from the intracellular store site in mesenteric resistance arteries of rats. It is speculated that mexiletine possesses greater inhibitory effects against noradrenaline-activated, verapamil-insensitive (receptor-operated) Ca2+ channels than against verapamil-sensitive (voltage-dependent) channels.
Collapse
Affiliation(s)
- Y Dohi
- Second Department of Internal Medicine, Nagoya City University Hospital, Japan
| | | | | |
Collapse
|
49
|
Abstract
This article reports a series of 31 patients with true phantom pain. Mexiletine was used in an open-label study to evaluate its effectiveness in the treatment of this disorder. Eighteen of these patients had good to excellent results with mexiletine alone. Eleven other patients responded favorably with a combination of mexiletine and clonidine. Two patients did not respond to either drug alone or in combination. Three patients elected to discontinue use of the mexiletine because of persistent nausea, even at low doses.
Collapse
Affiliation(s)
- R W Davis
- Pain Review, Inc., Colorado Springs, CO 80936
| |
Collapse
|
50
|
Bruno R, Santoni Y, Iliadis A, Djiane P, Serradimigni A, Cano JP. Simultaneous modelling of mexiletine and hydroxy-methyl-mexiletine data after single- and multiple-dose administration of a sustained-release mexiletine formulation. Biopharm Drug Dispos 1992; 13:481-93. [PMID: 1489940 DOI: 10.1002/bdd.2510130702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The pharmacokinetics of mexiletine and its metabolite hydroxy-methyl-mexiletine have been investigated following single-dose and during multiple-dose administration of a sustained-release form of mexiletine to six post-myocardial infarct patients. Comparison of single-dose and washout pharmacokinetics, after short-term multiple-dose administration, showed significant (p < 0.005), but not systematic, modifications in mexiletine apparent clearance for three patients. Furthermore, for these patients, simulation with both sets of parameters indicated that the steady-state was achieved before washout experiment in two cases. The fraction of mexiletine metabolized to hydroxy-methyl-mexiletine was lower for multiple-dose administration (about 18 per cent) than for the single dose (about 42 per cent). The hydroxy-methyl-mexiletine elimination rate constant was about four times that of mexiletine. Mexiletine clearance could be accounted for by other metabolic pathways. In one patient, hydroxy-methyl-mexiletine was undetectable even during multiple-dose administration, despite a significant increase in mexiletine clearance. However, the observed changes in mexiletine disposition had no therapeutic implications and active plasma levels were achieved by the third day of administration and maintained in the therapeutic range (0.75 to 2 micrograms ml-1) in all patients after a twice daily dosage regimen.
Collapse
Affiliation(s)
- R Bruno
- Laboratoire de Pharmacocinétique, INSERUM U278, Faculté de Pharmacie, Marseille, France
| | | | | | | | | | | |
Collapse
|