Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
PLoS One ; 19(5): e0304415, 2024.
Article in English | MEDLINE | ID: mdl-38820517

ABSTRACT

Fabry disease (FD) is an X-linked disorder of glycosphingolipid metabolism caused by mutations in the GLA gene encoding alpha-galactosidase A (α-Gal). Loss of α-Gal activity leads to progressive lysosomal accumulation of α-Gal substrate, predominately globotriaosylceramide (Gb3) and its deacylated derivative globotriaosylsphingosine (lyso-Gb3). FD manifestations include early onset neuropathic pain, gastrointestinal symptoms, and later onset life-threatening renal, cardiovascular and cerebrovascular disorders. Current treatments can preserve kidney function but are not very effective in preventing progression of cardiovascular pathology which remains the most common cause of premature death in FD patients. There is a significant need for a translational model that could be used for testing cardiac efficacy of new drugs. Two mouse models of FD have been developed. The α-Gal A-knockout (GlaKO) model is characterized by progressive tissue accumulation of Gb3 and lyso-Gb3 but does not develop any Fabry pathology besides mild peripheral neuropathy. Reports of minor cardiac function abnormalities in GlaKO model are inconsistent between different studies. Recently, G3Stg/GlaKO was generated by crossbreeding GlaKO with transgenic mice expressing human Gb3 synthase. G3Stg/GlaKO demonstrate higher tissue substrate accumulation and develop cellular and tissue pathologies. Functional renal pathology analogous to that found in early stages of FD has also been described in this model. The objective of this study is to characterize cardiac phenotype in GlaKO and G3Stg/GlaKO mice using echocardiography. Longitudinal assessments of cardiac wall thickness, mass and function were performed in GlaKO and wild-type (WT) littermate controls from 5-13 months of age. G3Stg/GlaKO and WT mice were assessed between 27-28 weeks of age due to their shortened lifespan. Several cardiomyopathy characteristics of early Fabry pathology were found in GlaKO mice, including mild cardiomegaly [up-to-25% increase in left ventricular (LV mass)] with no significant LV wall thickening. The LV internal diameter was significantly wider (up-to-24% increase at 9-months), when compared to the age-matched WT. In addition, there were significant increases in the end-systolic, end-diastolic volumes and stroke volume, suggesting volume overload. Significant reduction in Global longitudinal strain (GLS) measuring local myofiber contractility of the LV was also detected at 13-months. Similar GLS reduction was also reported in FD patients. Parameters such as ejection fraction, fractional shortening and cardiac output were either only slightly affected or were not different from controls. On the other hand, some of the cardiac findings in G3Stg/GlaKO mice were inconsistent with Fabry cardiomyopathy seen in FD patients. This could be potentially an artifact of the Gb3 synthase overexpression under a strong ubiquitous promoter. In conclusion, GlaKO mouse model presents mild cardiomegaly, mild cardiac dysfunction, but significant cardiac volume overload and functional changes in GLS that can be used as translational biomarkers to determine cardiac efficacy of novel treatment modalities. The level of tissue Gb3 accumulation in G3Stg/GlaKO mouse more closely recapitulates the level of substrate accumulation in FD patients and may provide better translatability of the efficacy of new therapeutics in clearing pathological substrates from cardiac tissues. But interpretation of the effect of treatment on cardiac structure and function in this model should be approached with caution.


Subject(s)
Disease Models, Animal , Fabry Disease , Mice, Knockout , alpha-Galactosidase , Animals , Fabry Disease/genetics , Fabry Disease/complications , Fabry Disease/metabolism , Fabry Disease/pathology , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , Mice , Humans , Trihexosylceramides/metabolism , Male , Female
2.
Mol Ther Methods Clin Dev ; 29: 286-302, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37359415

ABSTRACT

Mucopolysaccharidosis II (MPS II) is a rare lysosomal storage disease characterized by deficient activity of iduronate-2-sulfatase (I2S), leading to pathological accumulation of glycosaminoglycans (GAGs) in tissues. We used iduronate-2-sulfatase knockout (Ids KO) mice to investigate if liver-directed recombinant adeno-associated virus vectors (rAAV8-LSP-hIDSco) encoding human I2S (hI2S) could cross-correct I2S deficiency in Ids KO mouse tissues, and we then assessed the translation of mouse data to non-human primates (NHPs). Treated mice showed sustained hepatic hI2S production, accompanied by normalized GAG levels in somatic tissues (including critical tissues such as heart and lung), indicating systemic cross-correction from liver-secreted hI2S. Brain GAG levels in Ids KO mice were lowered but not normalized; higher doses were required to see improvements in brain histology and neurobehavioral testing. rAAV8-LSP-hIDSco administration in NHPs resulted in sustained hepatic hI2S production and therapeutic hI2S levels in cross-corrected somatic tissues but no hI2S exposure in the central nervous system, perhaps owing to lower levels of liver transduction in NHPs than in mice. Overall, we demonstrate the ability of rAAV8-LSP-hIDSco to cross-correct I2S deficiency in mouse somatic tissues and highlight the importance of showing translatability of gene therapy data from rodents to NHPs, which is critical for supporting translation to clinical development.

3.
Drugs Context ; 122023.
Article in English | MEDLINE | ID: mdl-36876155

ABSTRACT

Background: Prucalopride is a selective serotonin type 4 (5-HT4) receptor agonist indicated for treatment of chronic idiopathic constipation (CIC) in adults (2 mg orally, daily). 5-HT4 receptors are present in the central nervous system; therefore, non-clinical and clinical assessments were performed to evaluate the tissue distribution and abuse potential of prucalopride. Methods: In vitro receptor-ligand binding studies were performed to assess the affinity of prucalopride (≤1 mM) for peptide receptors, ion channels, monoamine neurotransmitters and 5-HT receptors. The tissue distribution of 14C-prucalopride (5 mg base-equivalent/kg) was investigated in rats. Behavioural assessments in mice, rats and dogs after treatment with single or repeated (up to 24 months) subcutaneous or oral doses of prucalopride (0.02-640 mg/kg across species) were performed. Treatment-emergent adverse events possibly indicative of abuse potential during prucalopride CIC clinical trials were evaluated. Results: Prucalopride showed no appreciable affinity for the receptors and ion channels investigated; its affinity (at ≤100 µM) for other 5-HT receptors was 150-10,000 times lower than that for the 5-HT4 receptor. In rats, <0.1% of the administered dose was found in the brain and concentrations were below the limit of detection within 24 hours. At supratherapeutic doses (≥20 mg/kg), mice and rats exhibited palpebral ptosis, and dogs exhibited salivation, eyelid tremors, decubitis, pedalling movements and sedation. All clinical treatment-emergent adverse events, possibly indicative of abuse potential, except dizziness, occurred in <1% of patients treated with prucalopride or placebo. Conclusion: This series of non-clinical and clinical studies suggest low abuse potential for prucalopride.

4.
Am J Gastroenterol ; 118(6): 955-960, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36927957

ABSTRACT

Prokinetic agents, specifically 5-hydroxytryptamine type 4 (5-HT 4 ) receptor agonists, have been shown to provide relief in chronic idiopathic constipation (CIC). The first-generation 5-HT 4 agonists were initially withdrawn from use owing to associations with serious cardiovascular (CV) events. This review summarizes CV safety data for prucalopride, a high-affinity 5-HT 4 agonist approved in the United States in 2018 for adults with CIC. No significant effects of prucalopride on CV safety were observed in animal models or early-phase clinical studies, including a thorough QT study at therapeutic (2 mg) or supratherapeutic (10 mg) doses. Among 1,750 patients with CIC who received prucalopride (2-4 mg) in 5 phase 3 studies, no trends in CV adverse events, electrocardiogram parameters, or blood pressure were documented; ≤1.0%-2.0% of patients had prolonged QT interval corrected for heart rate (HR) using Fridericia formula after placebo or prucalopride treatment, and low HR occurred in ≤6.1% and ≤3.3% of these patients, respectively. In two 24-month observational studies among 2,468 patients, changes in electrocardiogram parameters over time were minor, except at occasional time points when significant changes from baseline were reported for HR or QT interval. In a real-world European CV safety study among 35,087 patients (prucalopride, 5,715; polyethylene glycol 3350 [PEG3350], 29,372), results were consistent for no evidence of increased risk of major adverse CV events among patients treated with prucalopride vs PEG3350 (incidence rate ratio = 0.64; 95% confidence interval 0.36-1.14). Studies to date have not raised concerns regarding the impact of prucalopride treatment on CV parameters.


Subject(s)
Laxatives , Serotonin , Humans , Laxatives/adverse effects , Serotonin/therapeutic use , Constipation/chemically induced , Constipation/drug therapy , Serotonin 5-HT4 Receptor Agonists/therapeutic use , Chronic Disease , Treatment Outcome
5.
Skelet Muscle ; 8(1): 34, 2018 10 27.
Article in English | MEDLINE | ID: mdl-30368252

ABSTRACT

BACKGROUND: Myostatin antagonists are being developed as therapies for Duchenne muscular dystrophy due to their strong hypertrophic effects on skeletal muscle. Engineered follistatin has the potential to combine the hypertrophy of myostatin antagonism with the anti-inflammatory and anti-fibrotic effects of activin A antagonism. METHODS: Engineered follistatin was administered to C57BL/6 mice for 4 weeks, and muscle mass and myofiber size was measured. In the mdx model, engineered follistatin was dosed for 12 weeks in two studies comparing to an Fc fusion of the activin IIB receptor or an anti-myostatin antibody. Functional measurements of grip strength and tetanic force were combined with tissue analysis for markers of necrosis, inflammation, and fibrosis to evaluate improvement in dystrophic pathology. RESULTS: In wild-type and mdx mice, dose-dependent increases in muscle mass and quadriceps myofiber size were observed for engineered follistatin. In mdx, increases in grip strength and tetanic force were combined with improvements in muscle markers for necrosis, inflammation, and fibrosis. Improvements in dystrophic pathology were greater for engineered follistatin than the anti-myostatin antibody. CONCLUSIONS: Engineered follistatin generated hypertrophy and anti-fibrotic effects in the mdx model.


Subject(s)
Activins/antagonists & inhibitors , Follistatin/therapeutic use , Muscular Dystrophies/drug therapy , Myostatin/antagonists & inhibitors , Animals , Follistatin/administration & dosage , Hand Strength , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
6.
Antiviral Res ; 157: 128-133, 2018 09.
Article in English | MEDLINE | ID: mdl-30040968

ABSTRACT

The human cytomegalovirus (CMV) UL97 kinase inhibitor maribavir is in Phase III clinical trials as antiviral therapy, including use for infections refractory or resistant to standard therapy. To assess its activity in combination with approved and experimental CMV antivirals, and with the mTor inhibitor rapamycin (sirolimus), drug effects were tested by in vitro checkerboard assays and the data were analyzed using a three dimensional model based on an independent effects definition of additive interactions. Baseline virus and representative drug-resistant mutants were tested. According to the volume of synergy at 95% confidence, maribavir showed additive interactions with foscarnet, cidofovir, letermovir and GW275175X when tested against wild type and mutant viruses, strong antagonism with ganciclovir, and strong synergy with rapamycin, the latter suggesting a potentially useful therapeutic combination.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Cytomegalovirus/drug effects , Drug Interactions , Ribonucleosides/pharmacology , Cell Line , Humans , Microbial Sensitivity Tests
7.
J Pharmacol Exp Ther ; 364(2): 156-169, 2018 02.
Article in English | MEDLINE | ID: mdl-29180358

ABSTRACT

Patients with chronic constipation benefit from treatment with 5-hydroxytryptamine 4 (5-HT4) receptor agonists. However, the first-generation 5-HT4 receptor agonists cisapride and tegaserod were withdrawn from the market owing to rare cardiovascular adverse events that were not 5-HT4-receptor-related but due to the lack of selectivity of these drugs. Here we report the nonclinical cardiovascular profile of the selective 5-HT4 receptor agonist prucalopride. To assess its non-5-HT4 receptor-mediated effects on cardiovascular electrophysiological parameters, in vitro studies were performed in human ether-à-go-go-related gene-transfected cells, guinea pig ventricular myocytes and papillary muscle preparations, rabbit and dog Purkinje fibers, and the Langendorff rabbit heart. In vivo experiments were performed in a rabbit model for drug-induced proarrhythmogenesis, in anesthetized guinea pigs, and anesthetized and conscious dogs. In addition, human platelet aggregation and coronary artery contraction were studied to exclude interactions that have been suggested to mediate the cardiovascular effects of tegaserod. Effects at 5-HT4 receptors were evaluated in piglet and human atrial myocardium, and in anesthetized pigs. Finally, cardiovascular endpoints were investigated in chronic, repeated-dose toxicology studies at very high prucalopride doses in rats and dogs. No relevant effects were observed in any of the cardiovascular studies at concentrations at least 50 times the therapeutic plasma level. Only in pigs were minor and transient increases in heart rate and blood pressure noted upon first exposure to prucalopride, at plasma levels at least 10 times higher than human therapeutic plasma levels. Prucalopride may thus provide therapeutic benefit without the cardiovascular risks reported for other 5-HT4 receptor agonists.


Subject(s)
Benzofurans/pharmacology , Cardiovascular System/drug effects , Receptors, Serotonin, 5-HT4/metabolism , Serotonin 5-HT4 Receptor Agonists/pharmacology , Animals , Dogs , Dose-Response Relationship, Drug , Electrophysiological Phenomena/drug effects , Guinea Pigs , HEK293 Cells , Heart Atria/cytology , Heart Rate/drug effects , Humans , Myocardial Contraction/drug effects , Myocardium/metabolism , Rabbits
8.
J Med Chem ; 57(23): 9796-810, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25389560

ABSTRACT

The optimization of a series of aminooxazoline xanthene inhibitors of ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is described. An early lead compound showed robust Aß lowering activity in a rat pharmacodynamic model, but advancement was precluded by a low therapeutic window to QTc prolongation in cardiovascular models consistent with in vitro activity on the hERG ion channel. While the introduction of polar groups was effective in reducing hERG binding affinity, this came at the expense of higher than desired Pgp-mediated efflux. A balance of low Pgp efflux and hERG activity was achieved by lowering the polar surface area of the P3 substituent while retaining polarity in the P2' side chain. The introduction of a fluorine in position 4 of the xanthene ring improved BACE1 potency (5-10-fold). The combination of these optimized fragments resulted in identification of compound 40, which showed robust Aß reduction in a rat pharmacodynamic model (78% Aß reduction in CSF at 10 mg/kg po) and also showed acceptable cardiovascular safety in vivo.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Oxazolone/analogs & derivatives , Protease Inhibitors/chemical synthesis , Xanthenes/chemical synthesis , Animals , Crystallography, X-Ray , HEK293 Cells , Humans , Inhibitory Concentration 50 , Male , Microsomes, Liver/metabolism , Oxazolone/chemical synthesis , Oxazolone/pharmacology , Rats, Sprague-Dawley , Structure-Activity Relationship , Xanthenes/pharmacology
9.
J Med Chem ; 57(23): 9811-31, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25363711

ABSTRACT

We have previously shown that the aminooxazoline xanthene scaffold can generate potent and orally efficacious BACE1 inhibitors although certain of these compounds exhibited potential hERG liabilities. In this article, we describe 4-aza substitution on the xanthene core as a means to increase BACE1 potency while reducing hERG binding affinity. Further optimization of the P3 and P2' side chains resulted in the identification of 42 (AMG-8718), a compound with a balanced profile of BACE1 potency, hERG binding affinity, and Pgp recognition. This compound produced robust and sustained reductions of CSF and brain Aß levels in a rat pharmacodynamic model and exhibited significantly reduced potential for QTc elongation in a cardiovascular safety model.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Benzopyrans/chemical synthesis , Protease Inhibitors/chemical synthesis , Pyridines/chemical synthesis , Spiro Compounds/chemical synthesis , Amyloid beta-Peptides/metabolism , Animals , Benzopyrans/pharmacology , Ether-A-Go-Go Potassium Channels/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Microsomes, Liver/metabolism , Pyridines/pharmacology , Rats, Sprague-Dawley , Spiro Compounds/pharmacology , Structure-Activity Relationship
10.
J Pharmacol Toxicol Methods ; 66(2): 84-91, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22613062

ABSTRACT

INTRODUCTION: Cardiac arrhythmia findings can be a challenge to interpret and difficult to attribute to background incidence or test article treatment. Thus, there is a growing need to better understand arrhythmia incidence in the experimental animal models used to assess the cardiovascular safety of new drugs. Currently, there is little information on the frequency of spontaneous cardiac arrhythmias in the cynomolgus monkey. METHODS: This study evaluated the baseline arrhythmia rate in a group (n=19) of non-naïve (drug-free) male telemetered cynomolgus monkeys at various timepoints over a 6 month period. When sampled, data were collected continuously (24 hour bins over a 6 month period) and the ECG waveforms analyzed for arrhythmia using a semi-automated approach with pattern recognition software. The arrhythmia data were evaluated to detect atrial and ventricular patterns, as well as changes associated with circadian rhythm. RESULTS: Evaluation of this data showed that cynomolgus monkeys can exhibit spontaneous arrhythmias (day cycle; means) of the following types: supraventricular premature contraction (SPC, 10.7%); escape beats (EB, 3.8%); and sinus node pause (SNP, 2.8%), with others below 2%. From the ventricular perspective, ventricular premature beats (VPB, 25.4% (day cycle mean)) were the most prevalent. Circadian analysis indicated that some arrhythmias had higher incidence during the night cycle: SNP (32.6%); EB (18.5%); atrioventricular block (AVB, 2.7%), an indication that the arrhythmia pattern is influenced by the diurnal cycle. DISCUSSION: Overall, the data demonstrated that a variety of spontaneous arrhythmias occur at low frequency in non-treated animals, and the incidence varies between animals, and within the same animal when repeatedly sampled. Given the low incidence in normal animals, continuous ECG sampling over multiple days is needed to establish an accurate arrhythmia "fingerprint" for each animal in dedicated telemetry colonies, which could assist the interpretation of arrhythmia findings that may occur in cardiovascular safety studies.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Macaca fascicularis/physiology , Telemetry/methods , Animals , Animals, Laboratory , Arrhythmias, Cardiac/physiopathology , Cardiac Complexes, Premature/physiopathology , Circadian Rhythm , Electroencephalography , Heart Block/physiopathology , Male , Models, Animal , Reference Values , Sinoatrial Node/physiopathology , Time Factors
11.
Br J Pharmacol ; 142(8): 1300-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277312

ABSTRACT

Ranolazine is a novel anti-ischemic drug that prolongs the QT interval. To evaluate the potential mechanisms and consequences, we studied: (i) Ranolazine's effects on HERG and IsK currents in Xenopus oocytes with two-electrode voltage clamp; (ii) effects of ranolazine, compared to d-sotalol, on effective refractory period (ERP), QT interval and ventricular rhythm in a dog model of acquired long QT syndrome; and (iii) effects on selected native currents in canine atrial myocytes with whole-cell patch-clamp technique. Ranolazine inhibited HERG and IsK currents with different potencies. HERG was inhibited with an IC(50) of 106 micromol l(-1), whereas the IC(50) for IsK was 1.7 mmol l(-1). d-Sotalol caused reverse use-dependent ERP and QT interval prolongation, whereas ranolazine produced modest, nonsignificant increases that plateaued at submaximal doses. Neither drug affected QRS duration. d-Sotalol had clear proarrhythmic effects, with all d-sotalol-treated dogs developing torsades de pointes (TdP) ventricular tachyarrhythmias, of which they ultimately died. In contrast, ranolazine did not generate TdP. Effects on I(Kr) and I(Ks) were similar to those on HERG and IsK. Ranolazine blocked I(Ca) with an IC(50) of approximately 300 micromol l(-1). I(Na) was unaffected. We conclude that ranolazine inhibits I(Kr) by blocking HERG currents, inhibits I(Ca) at slightly larger concentrations, and has modest and self-limited effects on the QT interval. Unlike d-sotalol, ranolazine does not cause TdP in a dog model. The greater safety of ranolazine may be due to its ability to inhibit I(Ca) at concentrations only slightly larger than those that inhibit I(Kr), thus producing offsetting effects on repolarization.


Subject(s)
Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Piperazines/pharmacology , Acetanilides , Animals , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/physiology , Dogs , Dose-Response Relationship, Drug , Electrophysiology , Ether-A-Go-Go Potassium Channels , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/physiology , Ranolazine , Xenopus
12.
Circulation ; 107(14): 1930-6, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12668526

ABSTRACT

BACKGROUND: Coronary artery disease is a significant risk factor for atrial fibrillation (AF), but the basis for this association is incompletely understood. The present study evaluated the hypothesis that atrial ischemia can create a substrate for AF maintenance. METHODS AND RESULTS: Atrial ischemia was induced by occlusion of an atrial arterial branch that did not provide blood flow to the ventricles. Atrial-arterial occlusion increased the duration of AF induced by burst pacing from 57+/-32 seconds (control) to 803+/-214 seconds (P<0.001) after 0.5 hour of occlusion and to 887+/-209 seconds (P<0.001) after 3 hours of occlusion. Prolonged AF (>20 minutes) was induced in 0 of 16 dogs (0%) under control conditions, 7 of 16 (44%, P<0.01) at 0.5 to 3 hours, and 5 of 13 (38%, P<0.01) 3 to 5 hours after occlusion. Atrial conduction was slowed substantially within the ischemic zone: eg, conduction delay was 8+/-1 ms at a cycle length of 200 ms, control, versus 22+/-5 ms (P<0.01) after 0.5 hours and 27+/-5 ms (P<0.001) after 3 hours of ischemia. Refractoriness was initially unaffected but was prolonged 5 hours after occlusion. Phase-delay analysis and high-density mapping confirmed severe conduction slowing in the ischemic zone. Histological examination confirmed the location of ischemic regions and revealed extensive ischemia-induced necrosis at sites of conduction delay. CONCLUSIONS: Experimental atrial ischemia creates a substrate for AF maintenance, apparently by causing local conduction slowing that promotes reentry. These results suggest that atrial ischemia may significantly promote AF, and may be relevant to AF mechanisms in association with coronary artery disease.


Subject(s)
Atrial Fibrillation/etiology , Heart Atria , Myocardial Ischemia/complications , Animals , Arrhythmias, Cardiac/etiology , Atrial Fibrillation/physiopathology , Cardiac Pacing, Artificial , Dogs , Electrophysiology , Heart Atria/physiopathology , Kinetics , Myocardial Ischemia/pathology
13.
Pacing Clin Electrophysiol ; 26(3): 752-64, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12698678

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia requiring medical therapy, and present treatment modalities are inadequate. Over the past few years, we have learned a great deal about the phenomenon of electrical remodeling, by which rapid atrial activation leads to changes in atrial electrical properties that promote AF initiation and maintenance. This knowledge opens up the possibility that electrical remodeling may itself be a novel therapeutic target in AF. The present paper reviews what is known about the basic mechanisms of atrial electrical remodeling and then discusses the experimental and clinical evidence that remodeling can be prevented by drug therapy. Despite great potential value, the development of pharmacological interventions to prevent atrial electrical remodeling is still in its infancy.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/physiopathology , Heart Conduction System/physiopathology , Tachycardia/prevention & control , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antioxidants/pharmacology , Atrial Fibrillation/drug therapy , Calcium Channel Blockers/therapeutic use , Electrophysiology , Heart Atria/physiopathology , Humans , Potassium Channel Blockers/therapeutic use , Sodium Channel Blockers/therapeutic use , Tachycardia/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...