1
|
Trovato C, Longobardi S, Passini E, Beattie KA, Holmes M, Chaudhary KW, Rossman EI, Rodriguez B. In silico predictions of drug-induced changes in human cardiac contractility align with experimental recordings. Front Pharmacol 2025; 16:1500668. [PMID: 40166463 PMCID: PMC11955705 DOI: 10.3389/fphar.2025.1500668] [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: 09/23/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025] Open
Abstract
Drug-induced changes in cardiac contractility (inotropy) can lead to cardiotoxicity, a major cause of discontinuation in drug development. Preclinical approaches to assess cardiac inotropy are imperfect, with in vitro assays limited to stem cell-derived or adult human primary cardiomyocytes. Human mechanistic in silico modelling and simulations are already successfully applied for proarrhythmia prediction, contributing to cardiac safety assessment strategies in early drug development. In this study, we investigated their ability to predict drug-induced effects on cardiac inotropy. We considered a validation set of 28 neutral/negative inotropic and 13 positive inotropic reference compounds and simulated their effects on cell contractility via ion channel inhibition and perturbation of nine biomechanical modelling parameters, respectively. For each compound, a wide range of drug concentrations was simulated in an experimentally calibrated control population of 323 human ventricular in silico cells. Simulated biomarkers indicating drug-induced inotropic effects were compared with in vitro preclinical data from the literature. Computer simulations predicted drug-induced inotropic changes observed in vitro for 25 neutral/negative inotropes and 10 positive inotropes. Predictions of negative inotropic changes were quantitatively in agreement for 86% of tested drugs. Active tension peak was identified as the biomarker with highest predictive potential. This study describes the validation and application of an in silico cardiac electromechanical model for drug safety evaluation, combining ion channel inhibition data and information on potential inotropic mechanisms to predict inotropic changes. Furthermore, a route for its integration as part of a preclinical drug safety assessment strategy is outlined.
Collapse
Affiliation(s)
- Cristian Trovato
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
- Systems Medicine, Clinical Pharmacology and Safety Science, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Stefano Longobardi
- Non-Clinical Safety, Pre-Clinical Sciences, GlaxoSmithKline, Stevenage, United Kingdom
| | - Elisa Passini
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Kylie A. Beattie
- Non-Clinical Safety, Pre-Clinical Sciences, GlaxoSmithKline, Stevenage, United Kingdom
| | - Maxx Holmes
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Khuram W. Chaudhary
- Non-Clinical Safety, Pre-Clinical Sciences, GlaxoSmithKline, Upper Providence, Collegeville, PA, United States
| | - Eric I. Rossman
- Non-Clinical Safety, Pre-Clinical Sciences, GlaxoSmithKline, Upper Providence, Collegeville, PA, United States
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
2
|
Mar PL, Horbal P, Chung MK, Dukes JW, Ezekowitz M, Lakkireddy D, Lip GYH, Miletello M, Noseworthy PA, Reiffel JA, Tisdale JE, Olshansky B, Gopinathannair R. Drug Interactions Affecting Antiarrhythmic Drug Use. Circ Arrhythm Electrophysiol 2022; 15:e007955. [PMID: 35491871 DOI: 10.1161/circep.121.007955] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antiarrhythmic drugs (AAD) play an important role in the management of arrhythmias. Drug interactions involving AAD are common in clinical practice. As AADs have a narrow therapeutic window, both pharmacokinetic as well as pharmacodynamic interactions involving AAD can result in serious adverse drug reactions ranging from arrhythmia recurrence, failure of device-based therapy, and heart failure, to death. Pharmacokinetic drug interactions frequently involve the inhibition of key metabolic pathways, resulting in accumulation of a substrate drug. Additionally, over the past 2 decades, the P-gp (permeability glycoprotein) has been increasingly cited as a significant source of drug interactions. Pharmacodynamic drug interactions involving AADs commonly involve additive QT prolongation. Amiodarone, quinidine, and dofetilide are AADs with numerous and clinically significant drug interactions. Recent studies have also demonstrated increased morbidity and mortality with the use of digoxin and other AAD which interact with P-gp. QT prolongation is an important pharmacodynamic interaction involving mainly Vaughan-Williams class III AAD as many commonly used drug classes, such as macrolide antibiotics, fluoroquinolone antibiotics, antipsychotics, and antiemetics prolong the QT interval. Whenever possible, serious drug-drug interactions involving AAD should be avoided. If unavoidable, patients will require closer monitoring and the concomitant use of interacting agents should be minimized. Increasing awareness of drug interactions among clinicians will significantly improve patient safety for patients with arrhythmias.
Collapse
Affiliation(s)
- Philip L Mar
- Department of Medicine, Division of Cardiology, St. Louis University, St. Louis, MO (P.L.M., P.H.)
| | - Piotr Horbal
- Department of Medicine, Division of Cardiology, St. Louis University, St. Louis, MO (P.L.M., P.H.)
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute (M.K.C.), Cleveland Clinic, OH
| | | | - Michael Ezekowitz
- Lankenau Heart Institute, Bryn Mawr Hospital & Sidney Kimmel Medical College (M.E.)
| | | | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool & Liverpool Heart & Chest Hospital, Liverpool, United Kingdom (G.Y.H.L.).,Department of Clinical Medicine, Aalborg, Denmark (G.Y.H.L.)
| | | | - Peter A Noseworthy
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (P.A.N.)
| | - James A Reiffel
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.)
| | - James E Tisdale
- College of Pharmacy, Purdue University (J.E.T.).,School of Medicine, Indiana University, Indianapolis (J.E.T.)
| | - Brian Olshansky
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City (B.O.)
| | | | | |
Collapse
|
3
|
Optogenetic manipulation of cardiac electrical dynamics using sub-threshold illumination: dissecting the role of cardiac alternans in terminating rapid rhythms. Basic Res Cardiol 2022; 117:25. [PMID: 35488105 PMCID: PMC9054908 DOI: 10.1007/s00395-022-00933-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/01/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023]
Abstract
Cardiac action potential (AP) shape and propagation are regulated by several key dynamic factors such as ion channel recovery and intracellular Ca2+ cycling. Experimental methods for manipulating AP electrical dynamics commonly use ion channel inhibitors that lack spatial and temporal specificity. In this work, we propose an approach based on optogenetics to manipulate cardiac electrical activity employing a light-modulated depolarizing current with intensities that are too low to elicit APs (sub-threshold illumination), but are sufficient to fine-tune AP electrical dynamics. We investigated the effects of sub-threshold illumination in isolated cardiomyocytes and whole hearts by using transgenic mice constitutively expressing a light-gated ion channel (channelrhodopsin-2, ChR2). We find that ChR2-mediated depolarizing current prolongs APs and reduces conduction velocity (CV) in a space-selective and reversible manner. Sub-threshold manipulation also affects the dynamics of cardiac electrical activity, increasing the magnitude of cardiac alternans. We used an optical system that uses real-time feedback control to generate re-entrant circuits with user-defined cycle lengths to explore the role of cardiac alternans in spontaneous termination of ventricular tachycardias (VTs). We demonstrate that VT stability significantly decreases during sub-threshold illumination primarily due to an increase in the amplitude of electrical oscillations, which implies that cardiac alternans may be beneficial in the context of self-termination of VT.
Collapse
|
4
|
Crystal Structures of Antiarrhythmic Drug Disopyramide and Its Salt with Phthalic Acid. CRYSTALS 2021. [DOI: 10.3390/cryst11040379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Disopyramide (DPA) is as a class IA antiarrhythmic drug and its crystallization from cyclohexane at ambient condition yields lower melting form crystals which belong to the monoclinic centrosymmetric space group P21/n, having two molecules in an asymmetric unit. Crystal structure analysis of pure DPA revealed closely associated DPA molecules aggregates via amide–amide dimer synthon through the N–H∙∙∙O hydrogen bond whereas the second amide hydrogen N–H engaged in an intramolecular N–H∙∙∙N hydrogen bond with N-nitrogen of 2-pyridine moieties. Crystallization of DPA and phthalic acid (PA) in 1: 1 stoichiometric molar ratio from acetone at ambient condition yielded block shape crystals of 1:1 DPA_PA salt. Its X-ray single crystal structure revealed the formation of salt by transfer of acidic proton from one of the carboxylic acidic groups of PA to the tertiary amino group of chain moiety (N3-nitrogen atom) of DPA molecules. DPA_PA salt crystals belong to the monoclinic centrosymmetric space group P21/n, comprising one protonated DPA and one PA¯ anion (hydrogen phthalate counterion) in an asymmetric unit and linked by N–H∙∙∙O and C–H∙∙∙O hydrogen bonds. Pure DPA and DPA_PA salt were further characterized by differential calorimetric analysis, thermal gravimetric analysis, powder x-ray diffraction and infrared spectroscopy.
Collapse
|
5
|
Kelly MA. Neurological complications of cardiovascular drugs. HANDBOOK OF CLINICAL NEUROLOGY 2021; 177:319-344. [PMID: 33632450 DOI: 10.1016/b978-0-12-819814-8.00020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cardiovascular drugs are used to treat arterial hypertension, hyperlipidemia, arrhythmias, heart failure, and coronary artery disease. They also include antiplatelet and anticoagulant drugs that are essential for prevention of cardiogenic embolism. Most neurologic complications of the cardiovascular drugs are minor or transient and are far outweighed by the anticipated benefits of treatment. Other neurologic complications are more serious and require early recognition and management. Overtreatment of arterial hypertension may cause lightheadedness or fatigue but often responds readily to dose adjustment or an alternative drug. Other drug complications may be more troublesome as in myalgia associated with statins or headache associated with vasodilators. The recognized bleeding risk of the antithrombotics requires careful calculation of risk/benefit ratios for individual patients. Many neurologic complications of cardiovascular drugs are well documented in clinical trials with known frequency and severity, but others are rare and recognized only in isolated case reports or small case series. This chapter draws on both sources to report the adverse effects on muscle, nerve, and brain associated with commonly used cardiovascular drugs.
Collapse
Affiliation(s)
- Michael A Kelly
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, United States.
| |
Collapse
|
6
|
Delaloye NJ, Tobler K, O'Neill T, Kotsakis A, Cooper J, Bank I, Gilfoyle E. Errors During Resuscitation: The Impact of Perceived Authority on Delivery of Care. J Patient Saf 2020; 16:73-78. [PMID: 28671912 DOI: 10.1097/pts.0000000000000359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to determine the influence of perceived authority on pediatric resuscitation teams' response to an incorrect order given by a medical superior. METHODS As part of a larger multicenter prospective interventional study, interprofessional pediatric resuscitation teams (n = 48) participated in a video-recorded simulated resuscitation scenario with an infant in unstable, refractory supraventricular tachycardia. A confederate actor playing a senior physician entered the scenario partway through and ordered the incorrect dose and delivery method of the antiarrhythmic, procainamide. Video recordings were analyzed with a modified Advocacy Inquiry Scale, assessing the teams' ability to challenge the incorrect order, and a novel confederate hierarchical demeanor rating. The association between Advocacy Inquiry score and hierarchical demeanor rating, and whether or not the confederate's incorrect order was followed were determined. RESULTS Fifty percent (n = 24) of resuscitation teams followed the confederate's incorrect order. The teams' ability to challenge the incorrect order (P < 0.0001) and confederate hierarchical demeanor rating (P < 0.05) were significantly associated with whether or not the incorrect order was followed. Significant differences between rates of following the incorrect order at different study sites were observed (P < 0.05). CONCLUSIONS The reluctance of resuscitation teams to appropriately challenge the incorrect order resulted in a high rate of inappropriate medication administration. The rate of teams following the incorrect order was significantly associated with poor challenging of the incorrect order and the hierarchical demeanor of the perceived authority figure. Institution-based factors may impact this rate of incorrect medication administration.
Collapse
Affiliation(s)
- Nicole Jane Delaloye
- From the Medical Education Specialization, Departments of Community Health Sciences
| | | | | | - Afrothite Kotsakis
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, Ontario
| | | | - Ilana Bank
- Division of Emergency Medicine, Department of Pediatrics, McGill University, Montreal, Quebec
| | - Elaine Gilfoyle
- Section of Critical Care Medicine, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
7
|
Manolis TA, Manolis AA, Apostolopoulos EJ, Papatheou D, Melita H, Manolis AS. Cardiac arrhythmias in pregnant women: need for mother and offspring protection. Curr Med Res Opin 2020; 36:1225-1243. [PMID: 32347120 DOI: 10.1080/03007995.2020.1762555] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac arrhythmias are the most common cardiac complication reported in pregnant women with and without structural heart disease (SHD); they are more frequent among women with SHD, such as cardiomyopathy and congenital heart disease (CHD). While older studies had indicated supraventricular tachycardia as the most common tachyarrhythmia in pregnancy, more recent data indicate an increase in the frequency of arrhythmias, with atrial fibrillation (AF) emerging as the most frequent arrhythmia in pregnancy, attributed to an increase in maternal age, cardiovascular risk factors and CHD in pregnancy. Importantly, the presence of any tachyarrhythmia during pregnancy may be associated with adverse maternal and fetal outcomes, including death. Thus, both the mother and the offspring need to be protected from such consequences. The use of antiarrhythmic drugs (AADs) depends on clinical presentation and on the presence of underlying SHD, which requires caution as it promotes pro-arrhythmia. In hemodynamically compromised women, electrical cardioversion is successful and safe to both mother and fetus. Use of beta-blockers appears quite safe; however, caution is advised when using other AADs, while no AAD should be used, if at all possible, during the first trimester when organogenesis takes place. Regarding the anticoagulation regimen in patients with AF, warfarin should be substituted with heparin during the first trimester, while direct oral anticoagulants are not indicated given the lack of data in pregnancy. Finally, for refractory arrhythmias, ablation and/or device implantation can be performed with current techniques in pregnant women, when needed, using minimal exposure to radiation. All these issues and relevant current guidelines are herein reviewed.
Collapse
|
8
|
So HC, Wong YH. Implications of de novo mutations in guiding drug discovery: A study of four neuropsychiatric disorders. J Psychiatr Res 2019; 110:83-92. [PMID: 30597425 DOI: 10.1016/j.jpsychires.2018.12.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/14/2018] [Accepted: 12/11/2018] [Indexed: 12/19/2022]
Abstract
Recent studies have suggested an important role of de novo mutations (DNMs) in neuropsychiatric disorders. As DNMs are not subject to elimination due to evolutionary pressure, they are likely to have greater disruptions on biological functions. While a number of sequencing studies have been performed on neuropsychiatric disorders, the implications of DNMs for drug discovery remain to be explored. In this study, we employed a gene-set analysis approach to address this issue. Four neuropsychiatric disorders were studied, including schizophrenia (SCZ), autistic spectrum disorders (ASD), intellectual disability (ID) and epilepsy. We first identified gene-sets associated with different drugs, and analyzed whether the gene-set pertaining to each drug overlaps with DNMs more than expected by chance. We also assessed which medication classes are enriched among the prioritized drugs. We discovered that neuropsychiatric drug classes were indeed significantly enriched for DNMs of all four disorders; in particular, antipsychotics and antiepileptics were the most strongly enriched drug classes for SCZ and epilepsy respectively. Interestingly, we revealed enrichment of several unexpected drug classes, such as lipid-lowering agents for SCZ and anti-neoplastic agents. By inspecting individual hits, we also uncovered other interesting drug candidates or mechanisms (e.g. histone deacetylase inhibition and retinoid signaling) that might warrant further investigations. Taken together, this study provided evidence for the usefulness of DNMs in guiding drug discovery or repositioning.
Collapse
Affiliation(s)
- Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China; KIZ-CUHK Joint Laboratory of Bioresources, Molecular Research of Common Diseases, Kunming Zoology Institute of Zoology, China.
| | - Yui-Hang Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
9
|
|
10
|
Abstract
INTRODUCTION SK channels have functional importance in the cardiac atrium of many species, including humans. Pharmacological blockage of SK channels has been reported to be antiarrhythmic in animal models of atrial fibrillation; however, the exact antiarrhythmic mechanism of SK channel inhibition remains unclear. OBJECTIVES We speculated that together with a direct inhibition of repolarizing SK current, the previously observed depolarization of the atrial resting membrane potential (RMP) after SK channel inhibition reduces sodium channel availability, thereby prolonging the effective refractory period and slowing the conduction velocity (CV). We therefore aimed at elucidating these properties of SK channel inhibition and the underlying antiarrhythmic mechanisms using microelectrode action potential (AP) recordings and CV measurements in isolated rat atrium. Automated patch clamping and two-electrode voltage clamp were used to access INa and IK,ACh, respectively. RESULTS The SK channel inhibitor N-(pyridin-2-yl)-4-(pyridin-2-yl)thiazol-2-amine (ICA) exhibited antiarrhythmic effects. ICA prevented electrically induced runs of atrial fibrillation in the isolated right atrium and induced atrial postrepolarization refractoriness and depolarized RMP. Moreover, ICA (1-10 μM) was found to slow CV; however, because of a marked prolongation of effective refractory period, the calculated wavelength was increased. Furthermore, at increased pacing frequencies, SK channel inhibition by ICA (10-30 μM) demonstrated prominent depression of other sodium channel-dependent parameters. ICA did not inhibit IK,ACh, but at concentrations above 10 μM, ICA use dependently inhibited INa. CONCLUSIONS SK channel inhibition modulates multiple parameters of AP. It prolongs the AP duration and shifts the RMP towards more depolarized potentials through direct ISK block. This indirectly leads to sodium channel inhibition through accumulation of state dependently inactivated channels, which ultimately slows conduction and decreases excitability. However, a contribution from a direct sodium channel inhibition cannot be ruled. We here propose that the primary antiarrhythmic mechanism of SK channel inhibition is through direct potassium channel block and through indirect sodium channel inhibition.
Collapse
|
11
|
Jia J, Liu G, Zhang M, Lu Y, Lu C, Liu Y, Zheng H, Wang W, Gui Y, Yu C, Li S, Wang Y. Determination of the novel antiarrhythmic drug sulcardine sulfate in human plasma by liquid chromatography tandem mass spectrometry and its application in a clinical pharmacokinetic study. Biomed Chromatogr 2015; 30:1291-6. [PMID: 26715470 DOI: 10.1002/bmc.3681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/26/2015] [Accepted: 12/23/2015] [Indexed: 11/05/2022]
Abstract
Sulcardine sulfate (Sul), a novel antiarrhythmic agent, is currently in phase I and phase II clinical trials. To elucidate its clinical pharmacokinetic characteristics, a rapid and accurate liquid chromatography-tandem mass spectrometry (LC-MS/MS) method has been developed and validated for the quantification of Sul in human plasma. Plasma samples were precipitated by acetonitrile and isotope-labeled sulcardine was added as internal standard. The analysis was carried out on a Capcell Pak C18 MG III column (100 × 2.0 mm, 5 μm) with 0.1% formic acid in acetonitrile solution and water (17:83, v/v) as mobile phase. The linear range was 5.0-1000 ng/mL for Sul, with a lower limit of quantification of 5.0 ng/mL. The intra- and inter-batch CVs were within ±11.0% and the accuracies were 4.9-107.3%. Our method, for the first time, allows the rapid (only 3.0 min) and accurate quantification of Sul in human plasma. The method has been successfully applied in the pharmacokinetic study of Sul in a clinical trial following oral administration of Sul to healthy volunteers. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jingying Jia
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Gangyi Liu
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Mengqi Zhang
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Youli Lu
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Chuan Lu
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Yun Liu
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Hongcao Zheng
- Department of Cardiology, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Wei Wang
- Department of Cardiology, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Yuzhou Gui
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Chen Yu
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Shuijun Li
- Central Laboratory, Shanghai Xuhui Central Hospital and Shanghai Clinical Center, Chinese Academy of Science, 966 Middle Huaihai Road, Shanghai, 200031, China
| | - Yiping Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| |
Collapse
|
12
|
Abstract
Maintaining adequate tissue perfusion depends on a variety of factors, all of which can be influenced by xenobiotics (substances foreign to the body, including pharmaceuticals, chemicals, and natural compounds). Volume status, systemic vascular resistance, myocardial contractility, and cardiac rhythm all play a significant role in ensuring hemodynamic stability and proper cardiovascular function. Direct effects on the nervous system, the vasculature, or the heart itself as well as indirect metabolic effects may play a significant role in the development of cardiotoxicity. This article is dedicated to discussion of the disruption of cardiovascular physiology by xenobiotics.
Collapse
|
13
|
Siva S, Thulasidhasan J, Rajendiran N. Host-guest inclusion complex of propafenone hydrochloride with α- and β-cyclodextrins: spectral and molecular modeling studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2013; 115:559-567. [PMID: 23872014 DOI: 10.1016/j.saa.2013.06.079] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/08/2013] [Accepted: 06/19/2013] [Indexed: 06/02/2023]
Abstract
Host-guest inclusion complexes of cyclodextrins (CDs) with a potential cardiovascular drug propafenone hydrochloride (PFO), were prepared and characterized using absorption, fluorescence, time-resolved fluorescence, SEM, FT-IR, DSC, (1)H NMR, XRD and PM3 methods. The spectral studies suggested the phenyl ring along with carbonyl group is present inside of CD cavity. Solvent studies revealed that the normal Stokes shifted band originates from the locally excited state and the large Stokes shifted band occurs due to the emission from ICT. Nanosecond time-resolved studies indicated that PFO exhibits biexponential decay in water and triexponential decay in CD, indicating the formation of 1:1 inclusion complex. The results from solid state studies showed important modifications in the physicochemical properties of free PFO. The ΔH, ΔG and ΔS of the complexation process were determined and it was found that the complexation processes were spontaneous. Investigations of thermodynamic and electronic properties confirmed the stability of the inclusion complex.
Collapse
Affiliation(s)
- S Siva
- Department of Chemistry, Annamalai University, Annamalai Nagar, 608 002 Tamil Nadu, India
| | | | | |
Collapse
|
14
|
Antiarrhythmic properties of some 1,4-disubstituted piperazine derivatives with α1-adrenoceptor affinities. Eur J Pharmacol 2013; 720:237-46. [DOI: 10.1016/j.ejphar.2013.10.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 10/10/2013] [Accepted: 10/16/2013] [Indexed: 01/20/2023]
|
15
|
Sakr SA, El-Gamal EM. Effect of grapefruit juice on amiodarone induced nephrotoxicity in albino rats. Toxicol Ind Health 2013; 32:68-75. [DOI: 10.1177/0748233713498443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Amiodarone is a potent antiarrhythmic drug that is used to treat ventricular and supraventricular tachyarrhythmias. The present work studied the effect of amiodarone on the kidney of albino rats and the possible ameliorative role of grapefruit juice. Administration of amiodarone by gastric intubation (18 mg/kg body weight (b.w.), daily for 5 weeks) caused many histological alterations including intertubular leucocytic infiltrations, degeneration of the renal tubules, and atrophy of the glomeruli. Amiodarone caused marked elevation in serum creatinine and blood urea nitrogen. Histochemical examination of the renal tubules revealed depletion of glycogen and total proteins. Besides, animals administered with amiodarone showed an increase of apoptotic bands as detected by gel electrophoresis. Treating animals with amiodarone and grapefruit juice (27 ml/kg b.w.) caused an improvement in histological and histochemical appearance of the kidney together with decrease of serum creatinine and blood urea nitrogen. Moreover, the apoptosis was decreased. It is concluded from the obtained results that grapefruit juice ameliorates the nephrotoxicity of amiodarone in albino rats and this may be due to the potent antioxidant effects of its components.
Collapse
Affiliation(s)
- Saber A Sakr
- Department of Zoology, Faculty of Science, Menoufia University, Shebin El-kom, Egypt
| | - Ezz M El-Gamal
- Department of Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Bozeman WP, Ali K, Winslow JE. Long QT Syndrome Unmasked in an Adult Subject Presenting with Excited Delirium. J Emerg Med 2013; 44:e207-10. [DOI: 10.1016/j.jemermed.2012.02.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 12/01/2011] [Accepted: 02/25/2012] [Indexed: 10/28/2022]
|
17
|
Harmer A, Abi-Gerges N, Morton M, Pullen G, Valentin J, Pollard C. Validation of an in vitro contractility assay using canine ventricular myocytes. Toxicol Appl Pharmacol 2012; 260:162-72. [DOI: 10.1016/j.taap.2012.02.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 02/06/2012] [Accepted: 02/13/2012] [Indexed: 12/21/2022]
|
18
|
Challenges in Therapeutic Drug Monitoring of Digoxin and Other Anti-Arrhythmic Drugs. Ther Drug Monit 2012. [DOI: 10.1016/b978-0-12-385467-4.00011-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Pasquié JL, Thireau J, Davy JM, Le Guennec JY, Richard S. Médicaments anti-arythmiques : Présent et futur. ARCHIVES OF CARDIOVASCULAR DISEASES SUPPLEMENTS 2011. [DOI: 10.1016/s1878-6480(11)70394-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
20
|
Nishida A, Takizawa T, Matsumoto A, Miki T, Seino S, Nakaya H. Inhibition of ATP-sensitive K+ channels and L-type Ca2+ channels by amiodarone elicits contradictory effect on insulin secretion in MIN6 cells. J Pharmacol Sci 2011; 116:73-80. [PMID: 21512308 DOI: 10.1254/jphs.10294fp] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Some class I antiarrhythmic drugs induce a sporadic hypoglycemia by producing insulin secretion via inhibition of ATP-sensitive K(+) (K(ATP)) channels of pancreatic β-cells. It remains undetermined whether amiodarone produces insulin secretion by inhibiting K(ATP) channels. In this study, effects of amiodarone on K(ATP) channels, L-type Ca(2+) channel, membrane potential, and insulin secretion were examined and compared with those of quinidine in a β-cell line (MIN6). Amiodarone as well as quinidine inhibited the openings of the K(ATP) channel in a concentration-dependent manner without affecting its unitary amplitude in inside-out membrane patches of single MIN6 cells, and the IC(50) values were 0.24 and 4.9 µM, respectively. The L-type Ca(2+) current was also inhibited by amiodarone as well as quinidine in a concentration-dependent manner. Although glibenclamide (0.1 µM) or quinidine (10 µM) significantly potentiated the insulin secretion from MIN6 cells, amiodarone (1-30 µM) failed to increase insulin secretion. Amiodarone (30 µM) and nifedipine (10 µM) significantly inhibited the increase in insulin secretion produced by 0.1 µM glibenclamide. Amiodarone (30 µM) produced a gradual decrease of the membrane potential, but did not produce repetitive electrical activity in MIN6 cells. Glibenclamide (1 µM) produced a slow depolarization, followed by spiking activity which was inhibited by 30 µM amiodarone. Thus, amiodarone is unlikely to produce hypoglycemia in spite of potent inhibitory action on K(ATP) channels in insulin-secreting cells, possibly due to its Ca(2+) channel-blocking action.
Collapse
Affiliation(s)
- Atsushi Nishida
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Thireau J, Pasquié JL, Martel E, Le Guennec JY, Richard S. New drugs vs. old concepts: a fresh look at antiarrhythmics. Pharmacol Ther 2011; 132:125-45. [PMID: 21420430 DOI: 10.1016/j.pharmthera.2011.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 03/01/2011] [Indexed: 01/10/2023]
Abstract
Common arrhythmias, particularly atrial fibrillation (AF) and ventricular tachycardia/fibrillation (VT/VF) are a major public health concern. Classic antiarrhythmic (AA) drugs for AF are of limited effectiveness, and pose the risk of life-threatening VT/VF. For VT/VF, implantable cardiac defibrillators appear to be the unique, yet unsatisfactory, solution. Very few AA drugs have been successful in the last few decades, due to safety concerns or limited benefits in comparison to existing therapy. The Vaughan-Williams classification (one drug for one molecular target) appears too restrictive in light of current knowledge of molecular and cellular mechanisms. New AA drugs such as atrial-specific and/or multichannel blockers, upstream therapy and anti-remodeling drugs, are emerging. We focus on the cellular mechanisms related to abnormal Na⁺ and Ca²⁺ handling in AF, heart failure, and inherited arrhythmias, and on novel strategies aimed at normalizing ionic homeostasis. Drugs that prevent excessive Na⁺ entry (ranolazine) and aberrant diastolic Ca²⁺ release via the ryanodine receptor RyR2 (rycals, dantrolene, and flecainide) exhibit very interesting antiarrhythmic properties. These drugs act by normalizing, rather than blocking, channel activity. Ranolazine preferentially blocks abnormal persistent (vs. normal peak) Na⁺ currents, with minimal effects on normal channel function (cell excitability, and conduction). A similar "normalization" concept also applies to RyR2 stabilizers, which only prevent aberrant opening and diastolic Ca²⁺ leakage in diseased tissues, with no effect on normal function during systole. The different mechanisms of action of AA drugs may increase the therapeutic options available for the safe treatment of arrhythmias in a wide variety of pathophysiological situations.
Collapse
Affiliation(s)
- Jérôme Thireau
- Inserm U1046 Physiologie & Médecine Expérimentale du Cœur et des Muscles, Université Montpellier-1, Université Montpellier-2, 34295 Montpellier Cedex 5, France
| | | | | | | | | |
Collapse
|
22
|
Chen WH, Wang WY, Zhang J, Yang D, Wang YP. State-dependent blockade of human ether-a-go-go-related gene (hERG) K(+) channels by changrolin in stably transfected HEK293 cells. Acta Pharmacol Sin 2010; 31:915-22. [PMID: 20686516 DOI: 10.1038/aps.2010.84] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIM To study the effect of changrolin on the K(+) channels encoded by the human ether-a-go-go-related gene (hERG). METHODS hERG channels were heterologously stably expressed in human embryonic kidney 293 cells, and the hERG K(+) currents were recorded using a standard whole-cell patch-clamp technique. RESULTS Changrolin inhibited hERG channels in a concentration-dependent and reversible manner (IC(50)=18.23 mumol/L, 95% CI: 9.27-35.9 mumol/L; Hill coefficient=-0.9446). In addition, changrolin shifted the activation curve of hERG channels by 14.3+/-1.5 mV to more negative potentials (P<0.01, n=9) but did not significantly affect the steady-state inactivation of hERG (n=5, P>0.05). The relative block of hERG channels by changrolin was close to zero at the time point of channel opening by the depolarizing voltage step and quickly increased afterwards. The maximal block was achieved in the inactivated state, with no further development of the open channel block. In the "envelope of tails" experiments, the time constants of activation were found to be 287.8+/-46.2 ms and 174.2+/-18.4 ms, respectively, for the absence and presence of 30 mumol/L changrolin (P<0.05, n=7). The onset of inactivation was accelerated significantly by changrolin between -40 mV and +60 mV (P<0.05, n=7). CONCLUSION The results demonstrate that changrolin is a potent hERG blocker that preferentially binds to hERG channels in the open and inactivated states.
Collapse
|
23
|
Chang PM, Silka MJ, Moromisato DY, Bar-Cohen Y. Amiodarone Versus Procainamide for the Acute Treatment of Recurrent Supraventricular Tachycardia in Pediatric Patients. Circ Arrhythm Electrophysiol 2010; 3:134-40. [DOI: 10.1161/circep.109.901629] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Philip M. Chang
- From the Divisions of Cardiology (P.M.C., M.J.S., Y.B.-C.) and Critical Care Medicine (D.Y.M.), Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Michael J. Silka
- From the Divisions of Cardiology (P.M.C., M.J.S., Y.B.-C.) and Critical Care Medicine (D.Y.M.), Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - David Y. Moromisato
- From the Divisions of Cardiology (P.M.C., M.J.S., Y.B.-C.) and Critical Care Medicine (D.Y.M.), Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Yaniv Bar-Cohen
- From the Divisions of Cardiology (P.M.C., M.J.S., Y.B.-C.) and Critical Care Medicine (D.Y.M.), Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| |
Collapse
|
24
|
Komura S, Chinushi M, Furushima H, Hosaka Y, Izumi D, Iijima K, Watanabe H, Yagihara N, Aizawa Y. Efficacy of procainamide and lidocaine in terminating sustained monomorphic ventricular tachycardia. Circ J 2010; 74:864-9. [PMID: 20339190 DOI: 10.1253/circj.cj-09-0932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The efficacy of antiarrhythmic drugs in terminating sustained monomorphic ventricular tachycardia (SMVT) was assessed in a retrospective manner to provide a basis for recommending their use. METHODS AND RESULTS The 90 patients were included in this study to evaluate the efficacy to terminate SMVT using procainamide or lidocaine. All patients were alert and responsive. The mean systolic blood pressure was 91+/-25 mmHg (range, 40-150 mmHg). SMVT was diagnosed from ECG recordings and later in an electrophysiologic study. VTs with a cycle length of 329+/-55 and 324+/-61 ms were treated with the mean doses of 358+/-50 mg and 81+/-30 mg of procainamide and lidocaine and were terminated in 53/70 (75.7%) and in 7/20 (35.0%) respectively. The drugs were discontinued if there was no rise in blood pressure after slowing of the tachycardia rate or if there were signs of impending deterioration in consciousness. Though procainamide was effective, blood pressure was often low and DC shock should be available at all times during administration of the drug. CONCLUSIONS Procainamide, the relatively older drug, was more effective than lidocaine in terminating SMVT associated with structural heart diseases. This is a retrospective analysis but can form the basis for formulating guidelines for initial management of SMVT.
Collapse
Affiliation(s)
- Satoru Komura
- Division of Cardiology, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Guo L, Dong Z, Guthrie H. Validation of a guinea pig Langendorff heart model for assessing potential cardiovascular liability of drug candidates. J Pharmacol Toxicol Methods 2009; 60:130-51. [DOI: 10.1016/j.vascn.2009.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 07/06/2009] [Indexed: 02/02/2023]
|
26
|
Le Grand B, Pignier C, Létienne R, Colpaert F, Cuisiat F, Rolland F, Mas A, Borras M, Vacher B. Na+ Currents in Cardioprotection: Better to Be Late. J Med Chem 2009; 52:4149-60. [DOI: 10.1021/jm900296e] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
Wu L, Guo D, Li H, Hackett J, Yan GX, Jiao Z, Antzelevitch C, Shryock JC, Belardinelli L. Role of late sodium current in modulating the proarrhythmic and antiarrhythmic effects of quinidine. Heart Rhythm 2008; 5:1726-34. [PMID: 19084812 DOI: 10.1016/j.hrthm.2008.09.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 09/03/2008] [Indexed: 12/19/2022]
Abstract
BACKGROUND Quinidine is used to treat atrial fibrillation and ventricular arrhythmias. However, at low concentrations, it can induce torsade de pointes (TdP). OBJECTIVE The purpose of this study was to examine the role of late sodium current (I(Na)) as a modulator of the arrhythmogenicity of quinidine in female rabbit isolated hearts and cardiomyocytes. METHODS Epicardial and endocardial monophasic action potentials (MAPs), ECG signals, and ion channel currents were measured. The sea anemone toxin ATX-II was used to increase late I(Na). RESULTS Quinidine had concentration-dependent and often biphasic effects on measures of arrhythmogenicity. Quinidine increased the duration of epicardial MAP (MAPD(90)), QT interval, transmural dispersion of repolarization (TDR), and ventricular effective refractory period. Beat-to-beat variability of MAPD(90) (BVR), the interval from peak to end of the T wave (Tpeak-Tend) and index of Tpeak-Tend/QT interval were greater at 0.1 to 3 micromol/L than at 10-30 micromol/L quinidine. In the presence of 1 nmol/L ATX-II, quinidine caused significantly greater concentration-dependent and biphasic changes of Tpeak-Tend, TDR, BVR, and index of Tpeak-Tend/QT interval. Quinidine (1 micromol/L) induced TdP in 2 and 13 of 14 hearts in the absence and presence of ATX-II, respectively. Increases of BVR, index of Tpeak-Tend/QT interval, and Tpeak-Tend were associated with quinidine-induced TdP. Quinidine inhibited I(Kr), peak I(Na), and late I(Na) with IC(50)s of 4.5 +/- 0.3 micromol/L, 11.0 +/- 0.7 micromol/L, and 12.0 +/- 0.7 micromol/L. CONCLUSION Quinidine had biphasic proarrhythmic effects in the presence of ATX-II, suggesting that late I(Na) is a modulator of the arrhythmogenicity of quinidine. Enhancement of late I(Na) increased proarrhythmia caused by low but not high concentrations of quinidine.
Collapse
Affiliation(s)
- Lin Wu
- Pharmacological Sciences, CV Therapeutics, Inc., Palo Alto, California 94304, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Handler SM, Shirts BH, Perera S, Becich MJ, Castle NG, Hanlon JT. Frequency of laboratory monitoring of chronic medications administered to nursing facility residents: results of a national Internet-based study. THE CONSULTANT PHARMACIST : THE JOURNAL OF THE AMERICAN SOCIETY OF CONSULTANT PHARMACISTS 2008; 23:387-95. [PMID: 18540792 PMCID: PMC2597281 DOI: 10.4140/tcp.n.2008.387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To determine the minimal frequency of laboratory monitoring of 30 types of chronic medications or classes that are administered to nursing facility residents and are either listed under pharmacy services tag F329 (the tag for unnecessary medications), or have a narrow therapeutic index. DESIGN AND SETTING Cross-sectional, Internet-based survey. PARTICIPANTS National sample of 500 pharmacists, 500 nurse practitioners, and 327 physicians. MAIN OUTCOME MEASURE Minimal frequency of monitoring, recorded as an interval of 1, 3, 6, 9, or 12 months, for each of 35 laboratory parameters (e.g., serum drug level, complete blood count, liver function tests) for the 30 types of chronic medications or classes. Agreement was defined as having two or more of the three professional groups select the same minimal monitoring interval. RESULTS Overall, 116 professionals (20 pharmacists, 48 physicians, and 48 nurse practitioners) completed the survey. Most respondents were women (58.6% [68/116]), and most had worked in nursing facilities for > 5 years (66.4% [77/116]). Regarding minimal laboratory monitoring intervals, respondents reached agreement concerning 33 of 35 parameters. They selected three or six months as the minimum interval for 30 of 35 parameters (85.7%), one month as the minimum for two parameters, and 12 months as the minimum for one parameter. CONCLUSION The multidisciplinary panel agreed that most medications that were listed under the F329 tag or have a narrow therapeutic index should have laboratory monitoring every three or six months. The results can be used by nursing facility professionals to establish minimal laboratory monitoring parameters for chronic medications, which may potentially reduce the occurrence of adverse drug reactions.
Collapse
Affiliation(s)
- Steven M Handler
- Department of Medicine, Division of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Camm AJ. Safety considerations in the pharmacological management of atrial fibrillation. Int J Cardiol 2008; 127:299-306. [PMID: 18191470 DOI: 10.1016/j.ijcard.2007.11.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Accepted: 11/02/2007] [Indexed: 01/08/2023]
Abstract
The pharmacological management of atrial fibrillation (AF) requires careful consideration from a safety perspective. This article focuses primarily on maintenance therapy using antiarrhythmic drugs (AADs). The foremost safety issue for AADs is the propensity of class IA and III agents to cause torsade de pointes arrhythmias. Class IA drugs, particularly quinidine, can induce torsade de pointes at low or subtherapeutic doses, but higher doses are not necessarily associated with an increased incidence. 'Pure' class III drugs such as dofetilide induce torsade de pointes in a dose-related manner, but some class III agents with more complex actions such as amiodarone have a markedly lower potential to cause this arrhythmia. The risk of torsade de pointes precludes the use of class IA and 'pure' class III agents in patients with left ventricular hypertrophy and bradycardia. Class IC agents may cause sustained monomorphic ventricular tachycardias and are generally precluded in ischaemic and structural heart disease. Advanced heart failure patients may be treated with amiodarone or dofetilide, but most other AADs are unsuitable. The most important extracardiac toxicities occurring with AADs are those of amiodarone. Drug interactions are a significant safety issue in the management of AF, including pharmacokinetic interactions in which plasma levels of the AAD are raised - increasing the risk of proarrhythmia - and concomitant use of drugs that prolong the QT interval. Notwithstanding these considerations, most patients with AF can be considered for rhythm control, provided there is adequate pre-treatment assessment and protocols for initiation, dosing and monitoring are followed with care.
Collapse
Affiliation(s)
- A John Camm
- Department of Cardiological Sciences, St George's University of London, Cranmer Terrace, London SW17 0RE, United Kingdom.
| |
Collapse
|
30
|
|
31
|
Huston M, Levinson M. Are one or two dangerous? Quinine and quinidine exposure in toddlers. J Emerg Med 2007; 31:395-401. [PMID: 17046481 DOI: 10.1016/j.jemermed.2006.07.018] [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] [Received: 02/04/2005] [Revised: 08/11/2005] [Accepted: 01/13/2006] [Indexed: 10/24/2022]
Abstract
Quinine and quinidine have been cited as drugs that may cause significant morbidity and mortality in toddlers who ingest one or two pills. The use of both of these drugs has declined in the United States since the 1980s. A review of the literature and Poison Control data reveals that large quinine and quinidine ingestions, although rare in this country, may lead to severe toxicity and death related to cardiovascular and neurological effects in both children and adults. Although the majority of cases of quinine and quinidine toxicity in toddlers occur after ingestions of more than two pills, a single report each of severe toxicity after the equivalent of an ingestion of two pills or less by a toddler exists for both quinine and quinidine. Although the risk to the toddler exposed to one or two tablets seems to be small, triage to an Emergency Department is warranted after quinidine ingestion of any amount and after quinine ingestion that exceeds the age-appropriate therapeutic dose.
Collapse
Affiliation(s)
- Michelle Huston
- Department of Emergency Medicine, Franklin Square Hospital, 9000 Franklin Square Drive, Essex, MD 21237, USA
| | | |
Collapse
|
32
|
Lawrence CL, Bridgland-Taylor MH, Pollard CE, Hammond TG, Valentin JP. A rabbit Langendorff heart proarrhythmia model: predictive value for clinical identification of Torsades de Pointes. Br J Pharmacol 2006; 149:845-60. [PMID: 17031389 PMCID: PMC2014697 DOI: 10.1038/sj.bjp.0706894] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 06/19/2006] [Accepted: 08/11/2006] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE The rabbit isolated Langendorff heart model (SCREENIT) was used to investigate the proarrhythmic potential of a range of marketed drugs or drugs intended for market. These data were used to validate the SCREENIT model against clinical outcomes. EXPERIMENTAL APPROACH Fifty-five drugs, 3 replicates and 2 controls were tested in a blinded manner. Proarrhythmia variables included a 10% change in MAPD(60), triangulation, instability and reverse frequency-dependence of the MAP. Early after-depolarisations, ventricular tachycardia, TdP and ventricular fibrillation were noted. Data are reported at nominal concentrations relative to EFTPC(max). Proarrhythmic scores were assigned to each drug and each drug category. KEY RESULTS Category 1 and 2 drugs have the highest number of proarrhythmia variables and overt proarrhythmia while Category 5 drugs have the lowest, at every margin. At 30-fold the EFTPC(max), the mean proarrhythmic scores are: Category 1, 101+/-24; Category 2, 101+/-14; Category 3, 72+/-20; Category 4, 59+/-16 and Category 5, 22+/-9 points. Only drugs in Category 5 have mean proarrhythmic scores, below 30-fold, that remain within the Safety Zone. CONCLUSIONS AND IMPLICATIONS A 30-fold margin between effects and EFTPC(max) is sufficiently stringent to provide confidence to proceed with a new chemical entity, without incurring the risk of eliminating potentially beneficial drugs. The model is particularly useful where compounds have small margins between the hERG IC(50) and predicted EFTPC(max). These data suggest this is a robust and reliable assay that can add value to an integrated QT/TdP risk assessment.
Collapse
Affiliation(s)
- C L Lawrence
- Safety Pharmacology, Safety Assessment UK, AstraZeneca R&D, Macclesfield, Cheshire, UK.
| | | | | | | | | |
Collapse
|
33
|
Xie LH, Li Q, Lin AJ, Smith K, Zhang J, Skillman DS. New potential antimalarial agents: therapeutic-index evaluation of pyrroloquinazolinediamine and its prodrugs in a rat model of severe malaria. Antimicrob Agents Chemother 2006; 50:1649-55. [PMID: 16641431 PMCID: PMC1472225 DOI: 10.1128/aac.50.5.1649-1655.2006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tetra-acetamide pyrroloquinazolinediamine (PQD-A4) and bis-ethylcarbamyl pyrroloquinazolinediamine (PQD-BE) are new derivatives of pyrroloquinazolinediamine (PQD) and are being investigated as potential chemotherapeutic agents for the treatment of malaria. Comparative studies to assess the therapeutic indices of PQD-A4, PQD-BE, and PQD were conducted in Plasmodium berghei-infected rats following daily intragastric dosing for three consecutive days. Artesunate (AS), a standard drug for treatment of severe malaria, was used as a comparator. The minimum doses required to clear malaria parasitemia were 156 micromol/kg of body weight for AS and 2.4 micromol/kg for PQD, PQD-4A, and PQD-BE. The maximum tolerated dose (MTD) of AS was 625 micromol/kg, and its therapeutic index was calculated to be 4. The MTDs of PQD-A4, PQD-BE, and PQD were found to be 190, 77, and 24 micromol/kg, respectively, yielding therapeutic indices of 80, 32, and 10, respectively. Although PQD-A4 and PQD-BE are only half as potent as PQD based on their curative effects, the two new derivatives, PQD-4A and PQD-BE, are 8.0-fold and 3.2-fold safer, respectively, than their parent compound when they are dosed for three consecutive days. Oral PQD-A4 and PQD-BE are 44 to 70 times more potent on an mg basis than intravenous AS. As assessed from the therapeutic index over 3 days, PQD-A4, PQD-BE, and PQD administered orally are 20.0, 8.0, and 2.5 times safer than AS given intravenously. The results indicate that PQD-4A is a promising candidate for antimalarial treatment.
Collapse
Affiliation(s)
- Lisa H Xie
- Department of Pharmacology, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20307-5100, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Kalgutkar AS, Soglia JR. Minimising the potential for metabolic activation in drug discovery. Expert Opin Drug Metab Toxicol 2005; 1:91-142. [PMID: 16922655 DOI: 10.1517/17425255.1.1.91] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Investigations into the role of bioactivation in the pathogenesis of xenobiotic-induced toxicity have been a major area of research since the link between reactive metabolites and carcinogenesis was first reported in the 1930s. Circumstantial evidence suggests that bioactivation of relatively inert functional groups to reactive metabolites may contribute towards certain drug-induced adverse reactions. Reactive metabolites, if not detoxified, can covalently modify essential cellular targets. The identity of the susceptible biomacromolecule(s), and the physiological consequence of its covalent modification, will dictate the resulting toxicological response (e.g., covalent modification of DNA by reactive intermediates derived from procarcinogens that potentially leads to carcinogenesis). The formation of drug-protein adducts often carries a potential risk of clinical toxicities that may not be predicted from preclinical safety studies. Animal models used to reliably predict idiosyncratic drug toxicity are unavailable at present. Furthermore, considering that the frequency of occurrence of idiosyncratic adverse drug reactions (IADRs) is fairly rare (1 in 1000 to 1 in 10,000), it is impossible to detect such phenomena in early clinical trials. Thus, the occurrence of IADRs during late clinical trials or after a drug has been released can lead to an unanticipated restriction in its use and even in its withdrawal. Major themes explored in this review include a comprehensive cataloguing of bioactivation pathways of functional groups commonly utilised in drug design efforts with appropriate strategies towards detection of corresponding reactive intermediates. Several instances wherein replacement of putative structural alerts in drugs associated with IADRs with a latent functionality eliminates the underlying liability are also presented. Examples of where bioactivation phenomenon in drug candidates can be successfully abrogated via iterative chemical interventions are also discussed. Finally, appropriate strategies that aid in potentially mitigating the risk of IADRs are explored, especially in circumstances in which the structural alert is also responsible for the primary pharmacology of the drug candidate and cannot be replaced.
Collapse
Affiliation(s)
- Amit S Kalgutkar
- Pfizer Global Research & Development, Pharmacokinetics, Dynamics and Metabolism Department, Groton, CT 06340, USA.
| | | |
Collapse
|
35
|
Tsai CL. Quinidine cardiotoxicity. J Emerg Med 2005; 28:463-5. [PMID: 15837031 DOI: 10.1016/j.jemermed.2004.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2003] [Revised: 09/10/2004] [Accepted: 10/21/2004] [Indexed: 11/25/2022]
Affiliation(s)
- Chu-Lin Tsai
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
36
|
Ridley JM, Milnes JT, Benest AV, Masters JD, Witchel HJ, Hancox JC. Characterisation of recombinant HERG K+ channel blockade by the Class Ia antiarrhythmic drug procainamide. Biochem Biophys Res Commun 2003; 306:388-93. [PMID: 12804575 DOI: 10.1016/s0006-291x(03)00980-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Class Ia antiarrhythmic drugs, including procainamide (PROC), are associated with cardiac sodium channel blockade, delayed ventricular repolarisation and with a risk of ventricular pro-arrhythmia. The HERG K(+) channel is frequently linked to drug-induced pro-arrhythmia. Therefore, in this study, interactions between PROC and HERG K(+) channels were investigated, with particular reference to potency and mechanism of drug action. Whole-cell patch-clamp recordings of HERG current (I(HERG)) were made at 37 degrees C from human embryonic kidney (HEK 293) cells stably expressing the HERG channel. Following activating pulses to +20 mV, I(HERG) tails were inhibited by PROC with an IC(50) value of approximately 139 microM. I(HERG) blockade was found to be both time- and voltage-dependent, demonstrating contingency upon HERG channel gating. However, I(HERG) inhibition by PROC was relieved by depolarisation to a highly positive membrane potential (+80 mV) that favoured HERG channel inactivation. These data suggest that PROC inhibits the HERG K(+) channel by a primarily 'open' or 'activated' channel state blocking mechanism and that avidity of drug-binding is decreased by extensive I(HERG) inactivation. The potency of I(HERG) blockade by PROC is much lower than for other Class Ia agents that have been studied previously under analogous conditions (quinidine and disopyramide), although the blocking mechanism appears similar. Thus, differences between the chemical structure of PROC and other Class Ia antiarrhythmic drugs may help provide insight into chemical determinants of blocking potency for agents that bind to open/activated HERG channels.
Collapse
Affiliation(s)
- John M Ridley
- Department of Physiology and Cardiovascular Research Laboratories, School of Medical Sciences, University Walk, BS8 1TD, Bristol, UK
| | | | | | | | | | | |
Collapse
|
37
|
White SR, Dy G, Wilson JM. The case of the slandered Halloween cupcake: survival after massive pediatric procainamide overdose. Pediatr Emerg Care 2002; 18:185-8. [PMID: 12066005 DOI: 10.1097/00006565-200206000-00009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Suzanne R White
- Department of Emergency Medicine,Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | |
Collapse
|
38
|
Laven DL, Oller L. Drug Poisoning and Overdose for the Health Professional: Review of Select Over-the-Counter (OTC) and Prescription Medications. J Pharm Pract 2000. [DOI: 10.1177/089719000001300106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Exposure to over-the-counter and prescription medications can pose significant therapeutic and health hazards to patients, and present health care professionals with scenarios that require proper assessment and treatment. Knowing when an exposure to or overdose of a drug requires emergency medical attention is equally as important as to knowing when such assistance is not necessary—that simple treatment measures performed at home will suffice. This current discussion is intended to highlight select principles and clinical information pertaining to common drug exposures and overdoses, but not replace the full spectrum of information that would be available to health care professionals (and the lay public) by contacting their nearest poison control center. Many of the basic principles and concerns that are encountered with exposures to chemicals (i.e., route of exposure, patient medical history, quantity of the substance involved, elapsed time since the initial exposure, etc.) apply equally well to drug exposures. Likewise, evaluating each of these variables will determine which type of treatment approaches are, and are not, considered in situations of drug (or chemical) exposure and overdose.
Collapse
|
39
|
Anyukhovsky EP, Sosunov EA, Feinmark SJ, Rosen MR. Effects of quinidine on repolarization in canine epicardium, midmyocardium, and endocardium: II. In vivo study. Circulation 1997; 96:4019-26. [PMID: 9403627 DOI: 10.1161/01.cir.96.11.4019] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND In the companion article, we report a significant difference in quinidine effects on the action potential duration between surface (epicardial and endocardial) cells and midmyocardial cells (M cells) of canine left ventricle in vitro. This article considers two questions raised by the previous study: (1) Are the complex quinidine effects in vitro reflected in its actions on the heart in situ? (2) What are the cellular determinants of quinidine effects on QT interval in ECG? METHODS AND RESULTS We used plunge and surface electrodes to measure activation-recovery intervals (ARIs) of bipolar electrograms obtained from epicardium, endocardium, and midmyocardium (3, 5, and 9 mm from epicardium) of canine left ventricle in conditions of AV block and right ventricular pacing. Quinidine was infused continuously; its plasma level increased from 1.6+/-0.1 microg/mL at 30 minutes to 7.6+/-0.7 microg/mL at 180 minutes. At cycle lengths (CLs) from 300 to 1500 ms, there was no ARI gradient across the ventricular wall before and during quinidine infusion. At a CL of 300 ms, therapeutic concentrations of quinidine prolonged ARIs and QT intervals. At a CL of 1500 ms, ARIs were significantly prolonged at low quinidine concentrations. With an increase of quinidine concentration, this effect subsided and disappeared. CONCLUSIONS In situ, quinidine-induced prolongation of repolarization is uniform in all myocardial layers and follows the pattern observed in M cells in vitro. The ability of quinidine in therapeutic concentrations to prolong repolarization at rapid heart rates can contribute to its antiarrhythmic efficacy.
Collapse
Affiliation(s)
- E P Anyukhovsky
- Department of Pharmacology, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
40
|
Abstract
Poisoning by drugs that block voltage-gated sodium channels produces intraventricular conduction defects, myocardial depression, bradycardia, and ventricular arrhythmias. Human and animal reports suggest that hypertonic sodium bicarbonate may be effective therapy for numerous agents possessing sodium channel blocking properties, including cocaine, quinidine, procainamide, flecainide, mexiletine, bupivacaine, and others.
Collapse
Affiliation(s)
- P F Kolecki
- Department of Medical Toxicology, Good Samaritan Regional Medical Center, Phoenix, Arizona, USA
| | | |
Collapse
|
41
|
Brasch H. Activators of sodium, calcium and potassium channels modulate the cardiac effects of quinidine in vitro. PHARMACOLOGY & TOXICOLOGY 1995; 77:346-51. [PMID: 8778748 DOI: 10.1111/j.1600-0773.1995.tb01039.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Quinidine (25.5 mumol/l) reduced the beating frequency of isolated right guinea-pig atria, caused a negative inotropic effect in papillary muscles and slightly raised the contractile force of left atria. The functional refractory period of both tissues was prolonged. A 20% increase of the extracellular sodium concentration did not reverse the effects of quinidine. The Na-channel activator BDF 9148 (1 mumol/l) and the Ca-channel agonist Bay-K-8644 (0.5 mumol/l) further increased the contractile force and caused an additional prolongation of the functional refractory period in quinidine-pretreated atria. Only Bay-K-8644 was able to reverse the negative inotropic effect of quinidine in papillary muscles. The influence of Bay-K-8644 on the contractile force in quinidine-pretreated muscles was not attenuated by lemacalim (3 mumol/l), an activator of ATP-dependent potassium channels, but the duration of the functional refractory period was significantly reduced. These results suggest that a combination of a calcium channel activator and a potassium channel opener might be able to improve the treatment of quinidine intoxications.
Collapse
Affiliation(s)
- H Brasch
- Institute of Pharmacology, Medical University of Lübeck, Germany
| |
Collapse
|
42
|
Abstract
Cardiac dysfunction is often manifested as arrhythmia, with disruption of the normal periodicity and regularity of electromechanical activity. The therapy for arrhythmia begins with proper diagnosis, since many pharmacological interventions are themselves arrhythmogenic. Intervention for acute arrhythmia involves correction of underlying systemic conditions by ensuring adequate oxygenation, ventilation, acid-base homeostasis, electrolyte balance, and fluid status. Classification of antiarrhythmic agents assists in a structured treatment approach that utilizes different agents based on the etiology of the arrhythmia and the drug's mechanism of action. A deliberate treatment strategy guided by the morphological criteria of the arrhythmia modified by the rate and duration of complexes, noting symptoms and hemodynamic stability, is desirable.
Collapse
Affiliation(s)
- R B Vukmir
- Department of Anesthesiology/Critical Care and Emergency Medicine, University of Pittsburgh Medical Center, PA, USA
| |
Collapse
|
43
|
Leblanc M, Pichette V, Madore F, Ouimet D, Geadah D, Cardinal J. N-acetylprocainamide intoxication with torsade de pointes treated by high dialysate flow rate continuous arteriovenous hemodiafiltration. Crit Care Med 1995; 23:589-93. [PMID: 7533068 DOI: 10.1097/00003246-199503000-00027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- M Leblanc
- Service de Néphrologie, Hôpital Maisonneuve-Rosemont, Université de Montréal, PQ, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Kakei M, Nakazaki M, Kamisaki T, Nagayama I, Fukamachi Y, Tanaka H. Inhibition of the ATP-sensitive potassium channel by class I antiarrhythmic agent, cibenzoline, in rat pancreatic beta-cells. Br J Pharmacol 1993; 109:1226-31. [PMID: 8401933 PMCID: PMC2175729 DOI: 10.1111/j.1476-5381.1993.tb13753.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
1. Cibenzoline, a class I antiarrhythmic agent, was investigated for its effect on the ATP-sensitive K+ channel of pancreatic beta-cells by the patch clamp technique. 2. In perforated patch clamp experiments, cibenzoline depolarized the membrane of single beta-cells and thereafter, caused firing of action potentials in the presence of 2.8 mM glucose. 3. Cibenzoline inhibited the activity of the ATP-sensitive K+ channel in cell-attached recordings in the presence of 2.8 mM glucose and evoked repetitive fluctuations of the baseline current, apparently reflecting the action potentials of the beta-cell. 4. In whole-cell clamp experiments, time-independent outward current was induced by depleting cytoplasmic ATP with 0.1 mM ATP and 0.1 mM ADP in the solution contained in the pipette. The outward current was inhibited by cibenzoline in a dose-dependent manner in the concentration range of 1 microM to 100 microM and half maximum inhibition occurred at 1.5 microM. 5. Cibenzoline blocked substantially the ATP-sensitive K+ channel current when applied at the inner side of the membrane in isolated inside-out membrane patches. 6. It is concluded that cibenzoline blocks the ATP-sensitive K+ channel of pancreatic beta-cells and, thereby, stimulates insulin secretion at sub-stimulatory levels of glucose.
Collapse
Affiliation(s)
- M Kakei
- First Department of Internal Medicine, Faculty of Medicine, Kagoshima University, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Reiffel JA, Correia J. Evolutionary paths in arrhythmia management: influences of substrate, studies, and seismology. Am Heart J 1993; 125:1207-11. [PMID: 7682034 DOI: 10.1016/0002-8703(93)90151-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
46
|
|
47
|
Clark RF, Vance MV. Massive diphenhydramine poisoning resulting in a wide-complex tachycardia: successful treatment with sodium bicarbonate. Ann Emerg Med 1992; 21:318-21. [PMID: 1311158 DOI: 10.1016/s0196-0644(05)80897-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Diphenhydramine poisoning is characterized most often by anticholinergic effects. Cardiotoxicity and circulatory collapse have rarely been reported after massive ingestions of diphenhydramine and other H1-receptor-blocking agents, although these substances have local anesthetic properties and have been studied as antiarrhythmics. We report the case of a patient who developed a wide-complex tachycardia as a complication of acute diphenhydramine poisoning that responded to IV sodium bicarbonate.
Collapse
Affiliation(s)
- R F Clark
- Department of Medical Toxicology, Good Samaritan Regional Medical Center, Phoenix, Arizona
| | | |
Collapse
|