Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.146
1.
Cardiovasc Toxicol ; 24(5): 472-480, 2024 May.
Article En | MEDLINE | ID: mdl-38630336

The challenge posed by opioid overdose has become a significant concern for health systems due to the complexities associated with drug prohibition, widespread clinical use, and potential abuse. In response, healthcare professionals have primarily concentrated on mitigating the hallucinogenic and respiratory depressant consequences of opioid overdose to minimize associated risks. However, it is crucial to acknowledge that most opioids possess the capacity to prolong the QT interval, particularly in cases of overdose, thereby potentially resulting in severe ventricular arrhythmias and even sudden death if timely intervention is not implemented. Consequently, alongside addressing the typical adverse effects of opioids, it is imperative to consider their cardiotoxicity. To enhance comprehension of the correlation between opioids and arrhythmias, identify potential targets for prompt intervention, and mitigate the hazards associated with clinical utilization, an exploration of the interaction between drugs and ion channels, as well as their underlying mechanisms, becomes indispensable. This review primarily concentrates on elucidating the impact of opioid drugs on diverse ion channels, investigating recent advancements in this domain, and attaining a deeper understanding of the mechanisms underlying the prolongation of the QT interval by opioid drugs, along with potential interventions.


Analgesics, Opioid , Cardiotoxicity , Long QT Syndrome , Humans , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Analgesics, Opioid/adverse effects , Animals , Risk Assessment , Risk Factors , Heart Rate/drug effects , Action Potentials/drug effects , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Ion Channels/metabolism , Ion Channels/drug effects , Opiate Overdose/physiopathology
2.
J Pineal Res ; 73(1): e12798, 2022 Aug.
Article En | MEDLINE | ID: mdl-35384053

Melatonin treatment was reported to reduce the risk of cardiac arrhythmias, and crucial for this antiarrhythmic action was the effect of melatonin on activation spread. The aim of the present study was evaluation of the mechanisms of this activation enhancement. Experiments were performed in a total of 123 control and melatonin-treated (10 mg/kg, daily, for 7 days) male Wistar rats. In epicardial mapping studies (64 leads, interlead distance 0.5 mm) in the anesthetized animals, activation times (ATs) were determined in each lead as dV/dt minimum during QRS complex under sinus rhythm. Epicardial pacing was performed to measure conduction velocity (CV) across the mapped area. Average left ventricular ATs were shorter in the treated animals as compared to the controls, whereas the minimal epicardial ATs indicating the duration of activation propagation via the ventricular conduction system did not differ between the groups. CV was higher in the treated groups indicating that melatonin affected conduction via contractile myocardium The area of Cx43-derived fluorescence, as well as the expression of Cx43 protein, was similar in ventricles in the control and melatonin-treated groups. Expression of Gja1 gene transcripts encoding Cx43, was increased in the last group. An uncoupling agent octanol modified myocardial conduction properties (time of activation, action potential upstroke velocity, passive electrotonic phase duration) similarly in both groups. On the other hand, the expression of both Scn5a gene transcripts encoding Nav1.5 proteins, as well as peak density of transmembrane sodium current were increased in the ventricular myocytes from the melatonin-treated animals. Thus, a week-long melatonin treatment caused the increase of conduction velocity via enhancement of sodium channel proteins expression and increase of sodium current in the ventricular myocytes.


Connexin 43 , Heart Conduction System , Melatonin , NAV1.5 Voltage-Gated Sodium Channel , Animals , Connexin 43/genetics , Heart/physiology , Heart Conduction System/drug effects , Male , Melatonin/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Rats , Rats, Wistar , Sodium , Up-Regulation
3.
Cardiovasc Toxicol ; 22(1): 1-13, 2022 01.
Article En | MEDLINE | ID: mdl-33400130

Acute cardiovascular poisoning is a major cause of adverse outcomes in poisoning emergencies. The prognostic validity of corrected QT (QTc) and dispersed QT (QTd) in these outcomes is still limited. The present study aimed to determine the risk factors of mortality, adverse cardiovascular events (ACVE), and intensive care unit (ICU) admission in patients with acute cardiovascular toxicities and assess the validity of QTc and QTd intervals in predicting these outcomes. This study was conducted on adult patients admitted to Tanta University Poison Control Center with a history of acute cardiotoxic drugs or toxins exposure. The demographic and toxicological data of patients were recorded. Clinical examination, routine laboratory investigations, ECG grading, and measurement of QTc and QTd were performed. The patients were grouped according to their adverse outcomes. Among the included patients, 51 (31.48%) patients died, 61 (37.65%) patients had ACVE, and 68 (41.98%) patients required ICU admission. The most common cause of poisoning is aluminum phosphide, followed by cholinesterase inhibitors. QTd and QTdc showed no significant difference among outcome groups. The best cut-off values of QTc to predict mortality, ACVE, and ICU admission were > 491.1 ms, > 497.9 ms, and ≥ 491.9 ms, respectively. The derived cut-off QTc values were independent predictors for all adverse outcomes after adjusting for poison type, serum HCO3, and pulse. The highest odds ratios for all adverse outcomes were observed in aluminum phosphide poisoning and low HCO3 < 18 mmol/L. Thus, serum HCO3 and QTc interval should be monitored for acute cardiotoxicities, especially in aluminum phosphide and cholinesterase inhibitors poisoning.


Aluminum Compounds/poisoning , Arrhythmias, Cardiac/diagnosis , Cholinesterase Inhibitors/poisoning , Decision Support Techniques , Electrocardiography , Heart Conduction System/drug effects , Heart Rate/drug effects , Pesticides/toxicity , Phosphines/poisoning , Action Potentials , Adolescent , Adult , Aged , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/mortality , Arrhythmias, Cardiac/physiopathology , Cardiotoxicity , Egypt , Female , Heart Conduction System/physiopathology , Hospital Mortality , Humans , Intensive Care Units , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prospective Studies , Reproducibility of Results , Risk Assessment , Risk Factors , Time Factors , Young Adult
4.
Cardiovasc Res ; 118(2): 503-516, 2022 01 29.
Article En | MEDLINE | ID: mdl-33624748

AIMS: Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare laminopathy caused by expression of progerin, a lamin A variant, also present at low levels in non-HGPS individuals. HGPS patients age and die prematurely, predominantly from cardiovascular complications. Progerin-induced cardiac repolarization defects have been described previously, although the underlying mechanisms are unknown. METHODS AND RESULTS: We conducted studies in heart tissue from progerin-expressing LmnaG609G/G609G (G609G) mice, including microscopy, intracellular calcium dynamics, patch-clamping, in vivo magnetic resonance imaging, and electrocardiography. G609G mouse cardiomyocytes showed tubulin-cytoskeleton disorganization, t-tubular system disruption, sarcomere shortening, altered excitation-contraction coupling, and reductions in ventricular thickening and cardiac index. G609G mice exhibited severe bradycardia, and significant alterations of atrio-ventricular conduction and repolarization. Most importantly, 50% of G609G mice had altered heart rate variability, and sinoatrial block, both significant signs of premature cardiac aging. G609G cardiomyocytes had electrophysiological alterations, which resulted in an elevated action potential plateau and early afterdepolarization bursting, reflecting slower sodium current inactivation and long Ca+2 transient duration, which may also help explain the mild QT prolongation in some HGPS patients. Chronic treatment with low-dose paclitaxel ameliorated structural and functional alterations in G609G hearts. CONCLUSIONS: Our results demonstrate that tubulin-cytoskeleton disorganization in progerin-expressing cardiomyocytes causes structural, cardiac conduction, and excitation-contraction coupling defects, all of which can be partially corrected by chronic treatment with low dose paclitaxel.


Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Cytoskeleton/drug effects , Excitation Contraction Coupling/drug effects , Heart Conduction System/drug effects , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Paclitaxel/pharmacology , Progeria/drug therapy , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Lamin Type A/genetics , Lamin Type A/metabolism , Male , Mice, Mutant Strains , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Progeria/genetics , Progeria/metabolism , Progeria/physiopathology , Refractory Period, Electrophysiological/drug effects , Swine , Swine, Miniature , Tubulin/metabolism
5.
Circ Arrhythm Electrophysiol ; 15(1): e010273, 2022 01.
Article En | MEDLINE | ID: mdl-34961335

Nonmedical use of prescription and nonprescription drugs is a worldwide epidemic, rapidly growing in magnitude with deaths because of overdose and chronic use. A vast majority of these drugs are stimulants that have various effects on the cardiovascular system including the cardiac rhythm. Drugs, like cocaine and methamphetamine, have measured effects on the conduction system and through several direct and indirect pathways, utilizing multiple second messenger systems, change the structural and electrical substrate of the heart, thereby promoting cardiac dysrhythmias. Substituted amphetamines and cocaine affect the expression and activation kinetics of multiple ion channels and calcium signaling proteins resulting in EKG changes, and atrial and ventricular brady and tachyarrhythmias. Preexisting conditions cause substrate changes in the heart, which decrease the threshold for such drug-induced cardiac arrhythmias. The treatment of cardiac arrhythmias in patients who take drugs of abuse may be specialized and will require an understanding of the unique underlying mechanisms and necessitates a multidisciplinary approach. The use of primary or secondary prevention defibrillators in drug abusers with chronic systolic heart failure is both sensitive and controversial. This review provides a broad overview of cardiac arrhythmias associated with stimulant substance abuse and their management.


Amphetamine-Related Disorders/complications , Amphetamines/adverse effects , Arrhythmias, Cardiac/chemically induced , Central Nervous System Stimulants/adverse effects , Cocaine-Related Disorders/complications , Cocaine/adverse effects , Heart Conduction System/drug effects , Heart Rate/drug effects , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/therapy , Calcium Signaling/drug effects , Cardiotoxicity , Heart Conduction System/physiopathology , Humans , Prognosis , Risk Assessment , Risk Factors
6.
Pharmacol Res Perspect ; 9(5): e00874, 2021 10.
Article En | MEDLINE | ID: mdl-34628720

This study examined the safety, tolerability, and pharmacokinetics (PK) of napabucasin in healthy Asian and non-Asian participants and investigated the potential for QT/QTc interval prolongation. This five-part (A-E) study proceeded in a stepwise manner, unless stopping criteria were met. Parts A-D were randomized, double-blind, placebo-controlled, and included healthy Asian male and female and non-Asian male participants. PK parameters were measured following single-dose napabucasin (80-1200 mg) in the fasted or fed state (Part D). Potential QT/QTc interval prolongation was assessed using digital 12-lead electrocardiogram (Parts B and C). Part E was open-label, and examined the PK of single-dose napabucasin (240-720 mg) in healthy non-Asian males. Safety and tolerability were measured in Parts A-E. Changes from baseline in the Fridericia-corrected QT interval (ΔQTcF) and other electrocardiogram parameters were analyzed using a linear mixed-effects model. Napabucasin was well-tolerated across the study (n = 70), and no serious adverse events or significant safety issues were reported when administered with or without food. The most frequent treatment-emergent adverse events were diarrhea and abdominal pain, and these were mild in severity. No prolongation of the QTcF interval was reported following single-dose napabucasin (240-1200 mg) and changes in other cardiac parameters were negligible. The PK profile of napabucasin was consistent with earlier studies. Single-dose napabucasin was tolerated in healthy male and female participants, and no significant safety (including no QTcF prolongation) or tolerability issues were identified, irrespective of food intake. Clinical studies of napabucasin in advanced cancers are ongoing.


Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Heart Conduction System/drug effects , Naphthoquinones/pharmacology , Abdominal Pain/chemically induced , Antineoplastic Agents/adverse effects , Asian People , Benzofurans/adverse effects , Diarrhea/chemically induced , Double-Blind Method , Electrocardiography , Female , Healthy Volunteers , Humans , Long QT Syndrome/chemically induced , Male , Naphthoquinones/adverse effects , Reactive Oxygen Species
7.
PLoS Med ; 18(9): e1003766, 2021 09.
Article En | MEDLINE | ID: mdl-34492005

BACKGROUND: Amodiaquine is a 4-aminoquinoline antimalarial similar to chloroquine that is used extensively for the treatment and prevention of malaria. Data on the cardiovascular effects of amodiaquine are scarce, although transient effects on cardiac electrophysiology (electrocardiographic QT interval prolongation and sinus bradycardia) have been observed. We conducted an individual patient data meta-analysis to characterise the cardiovascular effects of amodiaquine and thereby support development of risk minimisation measures to improve the safety of this important antimalarial. METHODS AND FINDINGS: Studies of amodiaquine for the treatment or prevention of malaria were identified from a systematic review. Heart rates and QT intervals with study-specific heart rate correction (QTcS) were compared within studies and individual patient data pooled for multivariable linear mixed effects regression. The meta-analysis included 2,681 patients from 4 randomised controlled trials evaluating artemisinin-based combination therapies (ACTs) containing amodiaquine (n = 725), lumefantrine (n = 499), piperaquine (n = 716), and pyronaridine (n = 566), as well as monotherapy with chloroquine (n = 175) for uncomplicated malaria. Amodiaquine prolonged QTcS (mean = 16.9 ms, 95% CI: 15.0 to 18.8) less than chloroquine (21.9 ms, 18.3 to 25.6, p = 0.0069) and piperaquine (19.2 ms, 15.8 to 20.5, p = 0.0495), but more than lumefantrine (5.6 ms, 2.9 to 8.2, p < 0.001) and pyronaridine (-1.2 ms, -3.6 to +1.3, p < 0.001). In individuals aged ≥12 years, amodiaquine reduced heart rate (mean reduction = 15.2 beats per minute [bpm], 95% CI: 13.4 to 17.0) more than piperaquine (10.5 bpm, 7.7 to 13.3, p = 0.0013), lumefantrine (9.3 bpm, 6.4 to 12.2, p < 0.001), pyronaridine (6.6 bpm, 4.0 to 9.3, p < 0.001), and chloroquine (5.9 bpm, 3.2 to 8.5, p < 0.001) and was associated with a higher risk of potentially symptomatic sinus bradycardia (≤50 bpm) than lumefantrine (risk difference: 14.8%, 95% CI: 5.4 to 24.3, p = 0.0021) and chloroquine (risk difference: 8.0%, 95% CI: 4.0 to 12.0, p < 0.001). The effect of amodiaquine on the heart rate of children aged <12 years compared with other antimalarials was not clinically significant. Study limitations include the unavailability of individual patient-level adverse event data for most included participants, but no serious complications were documented. CONCLUSIONS: While caution is advised in the use of amodiaquine in patients aged ≥12 years with concomitant use of heart rate-reducing medications, serious cardiac conduction disorders, or risk factors for torsade de pointes, there have been no serious cardiovascular events reported after amodiaquine in widespread use over 7 decades. Amodiaquine and structurally related antimalarials in the World Health Organization (WHO)-recommended dose regimens alone or in ACTs are safe for the treatment and prevention of malaria.


Amodiaquine/adverse effects , Antimalarials/adverse effects , Bradycardia/chemically induced , Heart Conduction System/drug effects , Heart Rate/drug effects , Long QT Syndrome/chemically induced , Adolescent , Adult , Bradycardia/diagnosis , Bradycardia/physiopathology , Cardiotoxicity , Child , Child, Preschool , Female , Heart Conduction System/physiopathology , Humans , Infant , Long QT Syndrome/diagnosis , Long QT Syndrome/physiopathology , Male , Middle Aged , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors , Young Adult
8.
Prog Cardiovasc Dis ; 66: 70-79, 2021.
Article En | MEDLINE | ID: mdl-34332662

Electrical storm is present when a cluster of ventricular arrhythmias (VAs) occurs within a short time frame. The most widely accepted definition is 3 or more episodes of VA within a 24-h period, although prognostic risk begins to rise when 2 or more events occur within 3months. Electrical storm often presents as a medical emergency in the form of recurrent implantable cardiac defibrillator (ICD) shocks, recurrent syncope in patients with no ICD or low cardiac output symptoms. Management often requires a multimodality approach including ICD management, pharmacologic therapy, catheter ablation and modulations of the autonomic nervous system. In this article, we review the definition, prognosis and management of electrical storm.


Anti-Arrhythmia Agents/therapeutic use , Autonomic Denervation , Cardiac Pacing, Artificial , Catheter Ablation , Extracorporeal Membrane Oxygenation , Tachycardia, Ventricular/therapy , Ventricular Fibrillation/therapy , Action Potentials/drug effects , Anti-Arrhythmia Agents/adverse effects , Autonomic Denervation/adverse effects , Autonomic Denervation/mortality , Cardiac Pacing, Artificial/adverse effects , Cardiac Pacing, Artificial/mortality , Catheter Ablation/adverse effects , Catheter Ablation/mortality , Clinical Decision-Making , Decision Support Techniques , Extracorporeal Membrane Oxygenation/adverse effects , Extracorporeal Membrane Oxygenation/mortality , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Heart Conduction System/surgery , Heart Rate/drug effects , Humans , Pacemaker, Artificial , Recurrence , Risk Assessment , Risk Factors , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/mortality , Tachycardia, Ventricular/physiopathology , Treatment Outcome , Ventricular Fibrillation/diagnosis , Ventricular Fibrillation/mortality , Ventricular Fibrillation/physiopathology
9.
Cardiovasc Toxicol ; 21(11): 936-946, 2021 11.
Article En | MEDLINE | ID: mdl-34339022

Doxorubicin (DOX) is used as an anticancer drug despite its several side effects, especially its irreversible impacts on cardiotoxicity. Coenzyme Q10 (Q10) as a powerful antioxidant and lisinopril (LIS) as an angiotensin-converting enzyme inhibitor seem to provide protection against DOX-induced cardiotoxicity. Therefore, this study aimed to assess the cardioprotective effects of Q10 and LIS against DOX-induced cardiotoxicity in rats. Adult male Sprague-Dawley rats were randomly assigned into the control, LIS, Q10, DOX, DOX + LIS, and DOX + Q10 groups. On day 21, ECG was recorded and the right ventricle was dissected for evaluation of catalase activity and malondialdehyde (MDA) concentration. Additionally, the left ventricle and the sinoatrial (SA) node were dissected to assess the stereological parameters. The results of ECG indicated bradycardia and increase in QRS duration and QT interval in the DOX group compared to the control group. Meanwhile, the total volumes of the left ventricle, myocytes, and microvessels and the number of cardiomyocyte nuclei decreased, whereas the total volume of the connective tissue and the mean volume of cardiomyocytes increased in the DOX group. On the other hand, the SA node and the connective tissue were enlarged, while the volume of the SA node nuclei was reduced in the DOX group. Besides, catalase activity was lower and MDA concentration was higher in the DOX-treated group. Q10 could recover most stereological parameters, catalase activity, and MDA concentration. LIS also prevented some stereological parameters and ECG changes and improved catalase activity and MDA concentration in the DOX group. The findings suggested that Q10 and LIS exerted cardioprotective effects against DOX-induced cardiac toxicity.


Electrocardiography , Heart Conduction System/drug effects , Heart Diseases/prevention & control , Heart Rate/drug effects , Lisinopril/pharmacology , Myocytes, Cardiac/drug effects , Ubiquinone/analogs & derivatives , Animals , Antibiotics, Antineoplastic , Cardiotoxicity , Catalase/metabolism , Disease Models, Animal , Doxorubicin , Heart Conduction System/physiopathology , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/physiopathology , Male , Malondialdehyde/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Ubiquinone/pharmacology
11.
Am J Physiol Heart Circ Physiol ; 321(2): H294-H305, 2021 08 01.
Article En | MEDLINE | ID: mdl-34142884

The etiology of ethanol-related congenital heart defects has been the focus of much study, but most research has concentrated on cellular and molecular mechanisms. We have shown with optical coherence tomography (OCT) that ethanol exposure led to increased retrograde flow and smaller atrioventricular (AV) cushions compared with controls. Since AV cushions play a role in patterning the conduction delay at the atrioventricular junction (AVJ), this study aims to investigate whether ethanol exposure alters the AVJ conduction in early looping hearts and whether this alteration is related to the decreased cushion size. Quail embryos were exposed to a single dose of ethanol at gastrulation, and Hamburger-Hamilton stage 19-20 hearts were dissected for imaging. Cardiac conduction was measured using an optical mapping microscope and we imaged the endocardial cushions using OCT. Our results showed that, compared with controls, ethanol-exposed embryos exhibited abnormally fast AVJ conduction and reduced cushion size. However, this increased conduction velocity (CV) did not strictly correlate with decreased cushion volume and thickness. By matching the CV map to the cushion-size map along the inflow heart tube, we found that the slowest conduction location was consistently at the atrial side of the AVJ, which had the thinner cushions, not at the thickest cushion location at the ventricular side as expected. Our findings reveal regional differences in the AVJ myocardium even at this early stage in heart development. These findings reveal the early steps leading to the heterogeneity and complexity of conduction at the mature AVJ, a site where arrhythmias can be initiated.NEW & NOTEWORTHY To the best of our knowledge, this is the first study investigating the impact of ethanol exposure on the early cardiac conduction system. Our results showed that ethanol-exposed embryos exhibited abnormally fast atrioventricular conduction. In addition, our findings, in CV measurements and endocardial cushion thickness, reveal regional differences in the AVJ myocardium even at this early stage in heart development, suggesting that the differentiation and maturation at this site are complex and warrant further studies.


Central Nervous System Depressants/pharmacology , Endocardial Cushions/drug effects , Ethanol/pharmacology , Heart Conduction System/drug effects , Animals , Embryo, Nonmammalian , Endocardial Cushions/diagnostic imaging , Endocardial Cushions/embryology , Gastrulation , Heart/diagnostic imaging , Heart/drug effects , Heart/embryology , Heart Conduction System/diagnostic imaging , Heart Conduction System/embryology , Quail , Tomography, Optical Coherence , Voltage-Sensitive Dye Imaging
13.
Cardiovasc Toxicol ; 21(9): 687-694, 2021 09.
Article En | MEDLINE | ID: mdl-34018126

Several medicines, including cancer therapies, are known to alter the electrophysiological function of ventricular myocytes resulting in abnormal prolongation and dispersion of ventricular repolarization (quantified by multi-lead QTc measurement). This effect could be amplified by other concomitant factors (e.g., combination with other drugs affecting the QT, and/or electrolyte abnormalities, such as especially hypokalemia, hypomagnesaemia, and hypocalcemia). Usually, this condition results in higher risk of torsade de point and other life-threatening arrhythmias, related to unrecognized unpaired cardiac ventricular repolarization reserve (VRR). Being VRR a dynamic phenomenon, QT prolongation might often not be identified during the 10-s standard 12-lead ECG recording at rest, leaving the patient at increased risk for life-threatening event. We report the case of a 49-year woman, undergoing tamoxifen therapy for breast cancer, which alteration of ventricular repolarization reserve, persisting also after correction of concomitant recurrent hypokalemia, was evidenced only after manual measurements of the corrected QT (QTc) interval from selected intervals of the 12-lead ECG Holter monitoring. This otherwise missed finding was fundamental to drive the discontinuation of tamoxifen, shifting to another "safer" therapeutic option, and to avoid the use of potentially arrhythmogenic antibiotics when treating a bilateral pneumonia in recent COVID-19.


Anti-Bacterial Agents/therapeutic use , Arrhythmias, Cardiac/diagnosis , Breast Neoplasms/drug therapy , COVID-19 Drug Treatment , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Electrocardiography , Estrogen Antagonists/adverse effects , Heart Conduction System/drug effects , Tamoxifen/adverse effects , Action Potentials , Anti-Bacterial Agents/adverse effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , COVID-19/complications , COVID-19/diagnosis , Drug Substitution , Female , Heart Conduction System/physiopathology , Heart Rate/drug effects , Humans , Middle Aged , Predictive Value of Tests , Risk Assessment , Risk Factors
14.
Heart Rhythm ; 18(9): 1473-1479, 2021 09.
Article En | MEDLINE | ID: mdl-33932587

BACKGROUND: The optimal strategy for catheter ablation of persistent atrial fibrillation (PeAF) remains unknown. A preprocedural additive treatment for patients undergoing pulmonary vein isolation (PVI) alone to optimize catheter ablation should be investigated. OBJECTIVE: The purpose of this study was to determine whether pharmacologic cardioversion with a fixed low-dose antiarrhythmic drug (AAD) before ablation could stratify the long-term outcome of a PVI-alone strategy. METHODS: We conducted a prospective cohort study of PeAF patients who underwent PVI using contact force-sensing catheters. No substrate modification was performed. Fixed low-dose bepridil was administered before ablation for cardioversion and patients were classified into 2 groups based on obtaining sinus rhythm (SR). The rate of recurrence of atrial fibrillation (AF) and/or atrial tachycardia (AT) within 36 months was compared between the 2 groups. RESULTS: Among the 303 PeAF patients who received the AAD, 102 returned to SR (SR group), and the other 201 had persistence of AF (non-SR group). AF persistence duration at baseline and during bepridil administration was similar between the 2 groups. The SR group had a significantly lower 36-month AF/AT recurrence rate than the non-SR group (17 [22.2%] vs 55 [34.0%], log-rank P = .022). AT-type recurrence was observed in 16 patients (2 [3.3%] in the SR group vs 14 [8.9%] in the non-SR group; log-rank P = .051). Nonresponse to AAD was an independent predictor of AF/AT recurrence after adjusting for other risk factors (hazard ratio 1.34; 95% confidence interval 1.01-1.77; P = .040). CONCLUSION: Preprocedural pharmacologic cardioversion could be a useful determinant for patients with treatable PeAF by PVI alone.


Atrial Fibrillation/drug therapy , Bepridil/administration & dosage , Catheter Ablation , Heart Atria/physiopathology , Heart Conduction System/physiology , Preoperative Care/methods , Pulmonary Veins/surgery , Aged , Atrial Fibrillation/physiopathology , Atrial Fibrillation/surgery , Calcium Channel Blockers/administration & dosage , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Heart Conduction System/drug effects , Humans , Male , Prospective Studies , Recurrence , Risk Factors , Treatment Outcome
15.
Heart Rhythm ; 18(8): 1416-1422, 2021 08.
Article En | MEDLINE | ID: mdl-33848647

BACKGROUND: The negative inotropic effect of Class IC antiarrhythmic drugs limits their use for acute cardioversion of atrial fibrillation (AF). OBJECTIVE: The purpose of this study was to examine, in an intact porcine model, the effects of pulmonary and intravenous (IV) administration of flecainide on left ventricular (LV) contractility and QRS complex width at doses that are effective in converting new-onset AF to sinus rhythm. METHODS: Flecainide (1.5 mg/kg bolus) was delivered by intratracheal administration and compared to 2.0 mg/kg 10-minute IV administration (European Society of Cardiology guideline) and to 0.5 and 1.0 mg/kg 2-minute IV doses in 40 closed-chest, anesthetized Yorkshire pigs. Catheters were fluoroscopically positioned in the LV to monitor QRS complex width and contractility and at the bifurcation of the main bronchi to deliver intratracheal flecainide. RESULTS: Peak flecainide plasma concentrations (Cmax) were similar, but the 30-minute area under the curve (AUC) of plasma levels was 1.4- to 2.8-fold greater for 2.0 mg/kg 10-minute IV infusion than for the lower, more rapidly delivered intratracheal and IV doses. AUC for LV contractility (ie, negative inotropic burden) was 2.2- to 3.6-fold greater for 2.0 mg/kg 10-minute IV dose than for the lower, more rapidly delivered doses. QRS complex widening by flecainide was highly correlated with the decrease in LV contractility (r2 = 0.890, P <.0001, for all IV doses; r2 = 0.812, P = .01, for intratracheal flecainide). CONCLUSION: QRS complex widening in response to flecainide is strongly correlated with decrease in LV contractility. Rapid pulmonary or IV flecainide delivery reduces the negative inotropic burden while quickly achieving Cmax levels associated with conversion of AF.


Atrial Fibrillation/chemically induced , Electrocardiography , Flecainide/toxicity , Heart Conduction System/physiopathology , Heart Rate/physiology , Animals , Atrial Fibrillation/physiopathology , Disease Models, Animal , Heart Conduction System/drug effects , Heart Rate/drug effects , Male , Swine , Voltage-Gated Sodium Channel Blockers/toxicity
16.
Biomed Pharmacother ; 139: 111581, 2021 Jul.
Article En | MEDLINE | ID: mdl-33895523

Resibufogenin (RBG) is a chemical ingredient of Chan Su. In our research, we found RBG affected cardiac rhythm in a negative chronotropic way in vivo. The cardiac Mapping system ex vivo and the patch clamp in vitro were used to explore how RBG influenced the cardiac electrophysiological properties. The negative chronotropic action of RBG at 100 µM might be attribute to prolongation in the atrioventricular conduction time and reduction in the ventricular conduction velocity. Using whole-cell patch clamp in ventricular myocytes of adult rats, we found that RBG prolonged the action potential duration (APD) in APD20, APD50, and APD90 at 100 µM and inhibited calcium currents (ICa), total outward potassium currents (IK), and transient outward potassium current (Ito) in a concentration-dependent manner, but not on the inward rectifying potassium current (IK1). Notably, RBG had a potent proarrhythmic action ex vivo in the isolated perfused guinea pig hearts at 10 µM, but not in rats. To avoid the potential cardiotoxicity derived from the distributional differences of ion channels among species, the effect of RGB on IKr in hERG-HEK293 cells was detected. The IC50 of RGB on IKr was more than 100 µM. In summary, all these results indicated that the negative chronotropic action of RBG relied on the blocking activities on multiple ion channels, and the species-difference of proarrhythmic effects might result from lack of the Ito on the myocardial membrane of guinea pigs. Anyhow, the cardiotoxicity observed in guinea pigs required further detailed studies to mitigate the potential risks in the clinical application of Chan Su.


Bufanolides/pharmacology , Cardiotonic Agents/pharmacology , Heart Conduction System/drug effects , Heart/drug effects , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/chemically induced , Calcium Channels/drug effects , Electrocardiography/drug effects , Guinea Pigs , HEK293 Cells , Heart Rate/drug effects , Humans , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Potassium Channels/drug effects , Rats , Species Specificity
17.
Heart Vessels ; 36(7): 1088-1097, 2021 Jul.
Article En | MEDLINE | ID: mdl-33763729

To characterize in vivo anti-atrial fibrillatory potential and pharmacological safety profile of ranolazine having INa,L plus IKr inhibitory actions in comparison with those of clinically available anti-atrial fibrillatory drugs; namely, dronedarone, amiodarone, bepridil and dl-sotalol in our previous studies, ranolazine dihydrochloride in sub-therapeutic (0.3 mg/kg) and supra-therapeutic (3 mg/kg) doses was intravenously infused over 10 min to the halothane-anesthetized dogs (n = 5). The low dose increased the heart rate, cardiac output and atrioventricular conduction velocity possibly via vasodilator action-induced, reflex-mediated increase of adrenergic tone. Meanwhile, the high dose decreased the heart rate, ventricular contraction, cardiac output and mean blood pressure, indicating that drug-induced direct actions may exceed the reflex-mediated compensation. In addition, it prolonged the atrial and ventricular effective refractory periods, of which potency and selectivity for the former were less great compared with those of the clinically-available drugs. Moreover, it did not alter the ventricular early repolarization period in vivo, but prolonged the late repolarization with minimal risk for re-entrant arrhythmias. These in vivo findings of ranolazine suggest that INa,L suppression may attenuate IKr inhibition-associated prolongation of early repolarization in the presence of reflex-mediated increase of adrenergic tone. Thus, ranolazine alone may be less promising as an anti-atrial fibrillatory drug, but its potential risk for inducing torsade de pointes will be small. These information can be used as a guide to predict the utility and adverse effects of anti-atrial fibrillatory drugs having multi-channel modulatory action.


Anesthesia, Inhalation/methods , Atrial Fibrillation/drug therapy , Halothane/pharmacology , Heart Atria/physiopathology , Heart Conduction System/drug effects , Heart Rate/drug effects , Ranolazine/administration & dosage , Action Potentials/drug effects , Anesthetics, Inhalation/pharmacology , Animals , Atrial Fibrillation/physiopathology , Cardiac Output/drug effects , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Electrocardiography/drug effects , Female , Heart Atria/drug effects , Heart Conduction System/physiopathology , Infusions, Intravenous , Sodium Channel Blockers/administration & dosage
18.
Cardiovasc Res ; 117(7): 1616-1631, 2021 06 16.
Article En | MEDLINE | ID: mdl-33769493

Atrial fibrillation (AF) is the most common sustained clinical arrhythmia, with a lifetime incidence of up to 37%, and is a major contributor to population morbidity and mortality. Important components of AF management include control of cardiac rhythm, rate, and thromboembolic risk. In this narrative review article, we focus on rhythm-control therapy. The available therapies for cardiac rhythm control include antiarrhythmic drugs and catheter-based ablation procedures; both of these are presently neither optimally effective nor safe. In order to develop improved treatment options, it is necessary to use preclinical models, both to identify novel mechanism-based therapeutic targets and to test the effects of putative therapies before initiating clinical trials. Extensive research over the past 30 years has provided many insights into AF mechanisms that can be used to design new rhythm-maintenance approaches. However, it has proven very difficult to translate these mechanistic discoveries into clinically applicable safe and effective new therapies. The aim of this article is to explore the challenges that underlie this phenomenon. We begin by considering the basic problem of AF, including its clinical importance, the current therapeutic landscape, the drug development pipeline, and the notion of upstream therapy. We then discuss the currently available preclinical models of AF and their limitations, and move on to regulatory hurdles and considerations and then review industry concerns and strategies. Finally, we evaluate potential paths forward, attempting to derive insights from the developmental history of currently used approaches and suggesting possible paths for the future. While the introduction of successful conceptually innovative new treatments for AF control is proving extremely difficult, one significant breakthrough is likely to revolutionize both AF management and the therapeutic development landscape.


Action Potentials/drug effects , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Drug Development , Drug Discovery , Heart Conduction System/drug effects , Heart Rate/drug effects , Animals , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Diffusion of Innovation , Disease Models, Animal , Drug Approval , Drug Development/trends , Drug Discovery/trends , Forecasting , Heart Conduction System/physiopathology , Humans , Translational Research, Biomedical
19.
J Immunother Cancer ; 9(3)2021 03.
Article En | MEDLINE | ID: mdl-33653803

BACKGROUND: Myocarditis is a highly morbid complication of immune checkpoint inhibitor (ICI) use that remains inadequately characterized. The QRS duration and the QTc interval are standardized electrocardiographic measures that are prolonged in other cardiac conditions; however, there are no data on their utility in ICI myocarditis. METHODS: From an international registry, ECG parameters were compared between 140 myocarditis cases and 179 controls across multiple time points (pre-ICI, on ICI prior to myocarditis, and at the time of myocarditis). The association between ECG values and major adverse cardiac events (MACE) was also tested. RESULTS: Both the QRS duration and QTc interval were similar between cases and controls prior to myocarditis. When compared with controls on an ICI (93±19 ms) or to baseline prior to myocarditis (97±19 ms), the QRS duration prolonged with myocarditis (110±22 ms, p<0.001 and p=0.009, respectively). In contrast, the QTc interval at the time of myocarditis (435±39 ms) was not increased compared with pre-myocarditis baseline (422±27 ms, p=0.42). A prolonged QRS duration conferred an increased risk of subsequent MACE (HR 3.28, 95% CI 1.98 to 5.62, p<0.001). After adjustment, each 10 ms increase in the QRS duration conferred a 1.3-fold increase in the odds of MACE (95% CI 1.07 to 1.61, p=0.011). Conversely, there was no association between the QTc interval and MACE among men (HR 1.33, 95% CI 0.70 to 2.53, p=0.38) or women (HR 1.48, 95% CI 0.61 to 3.58, p=0.39). CONCLUSIONS: The QRS duration is increased in ICI myocarditis and is associated with increased MACE risk. Use of this widely available ECG parameter may aid in ICI myocarditis diagnosis and risk-stratification.


Action Potentials/drug effects , Electrocardiography , Heart Conduction System/drug effects , Heart Rate/drug effects , Immune Checkpoint Inhibitors/adverse effects , Myocarditis/diagnosis , Aged , Aged, 80 and over , Female , Heart Conduction System/physiopathology , Humans , Male , Middle Aged , Myocarditis/chemically induced , Myocarditis/physiopathology , Predictive Value of Tests , Registries , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors
20.
Cardiovasc Toxicol ; 21(4): 336-348, 2021 04.
Article En | MEDLINE | ID: mdl-33389603

Noise has become a prevalent public health problem across the world. Although there is a significant amount of data demonstrating the harmful effects of noise on the body, very little is known about how it impacts subsequent responses to other environmental stressors like air pollution, which tend to colocalize in urban centers. Therefore, this study was conducted to determine the effect of intermittent noise on cardiovascular function and subsequent responses to ozone (O3). Male Wistar-Kyoto rats implanted with radiotelemeters to non-invasively measure heart rate (HR) and blood pressure (BP), and assess heart rate variability (HRV) and baroreflex sensitivity (BRS) were kept in the quiet or exposed to intermittent white noise (85-90 dB) for one week and then exposed to either O3 (0.8 ppm) or filtered air. Left ventricular function and arrhythmia sensitivity were measured 24 h after exposure. Intermittent noise caused an initial increase in HR and BP, which decreased significantly later in the regimen and coincided with an increase in HRV and BRS. Noise caused HR and BP to be significantly elevated early during O3 and lower at the end when compared to animals kept in the quiet while the increased HRV and BRS persisted during the 24 h after. Lastly, noise increased arrhythmogenesis and may predispose the heart to mechanical function changes after O3. This is the first study to demonstrate that intermittent noise worsens the cardiovascular response to inhaled O3. These effects may occur due to autonomic changes and dysregulation of homeostatic controls, which persist one day after exposure to noise. Hence, co-exposure to noise should be taken into account when assessing the health effects of urban air pollution.


Arrhythmias, Cardiac/chemically induced , Heart Conduction System/drug effects , Heart Rate/drug effects , Noise/adverse effects , Ozone/toxicity , Animals , Arrhythmias, Cardiac/physiopathology , Baroreflex/drug effects , Blood Pressure/drug effects , Cardiotoxicity , Heart Conduction System/physiopathology , Inhalation Exposure/adverse effects , Male , Rats, Inbred WKY
...