Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 756
Filter
1.
Tex Heart Inst J ; 51(1)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38686681

ABSTRACT

BACKGROUND: Atrial tachyarrhythmias are common and difficult to treat in adults with congenital heart disease. Dronedarone has proven effective in patients without congenital heart disease, but data are limited about its use in adults with congenital heart disease of moderate to great complexity. METHODS: A single-center, retrospective chart review of 21 adults with congenital heart disease of moderate to great complexity who were treated with dronedarone for atrial tachyarrhythmias was performed. RESULTS: The median (IQR) age at dronedarone initiation was 35 (27.5-39) years. Eleven patients (52%) were male. Ten patients (48%) had New York Heart Association class I disease, 10 (48%) had class II disease, and 1 (5%) had class III disease. Ejection fraction at initiation was greater than 55% in 11 patients (52%), 35% to 55% in 9 patients (43%), and less than 35% in 1 patient (5%). Prior treatments included ß-blockers (71%), sotalol (38%), amiodarone (24%), digoxin (24%), and catheter ablation (38%). Rhythm control was complete in 5 patients (24%), partial in 6 (29%), and inadequate in 10 (48%). Two patients (10%) experienced adverse events, including nausea in 1 (5%) and cardiac arrest in 1 (5%), which occurred 48 months after initiation of treatment. There were no deaths during the follow-up period. The median (IQR) follow-up time for patients with complete or partial rhythm control was 20 (1-54) months. CONCLUSION: Dronedarone can be effective for adult patients with congenital heart disease and atrial arrhythmias for whom more established therapies have failed, and with close monitoring it can be safely tolerated.


Subject(s)
Anti-Arrhythmia Agents , Dronedarone , Heart Defects, Congenital , Humans , Dronedarone/therapeutic use , Dronedarone/adverse effects , Male , Retrospective Studies , Female , Adult , Heart Defects, Congenital/complications , Heart Defects, Congenital/physiopathology , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/adverse effects , Treatment Outcome , Heart Rate/drug effects , Heart Rate/physiology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Atrial Fibrillation/diagnosis , Atrial Fibrillation/complications , Amiodarone/therapeutic use , Amiodarone/adverse effects , Amiodarone/analogs & derivatives , Time Factors
2.
Biol Pharm Bull ; 45(7): 948-954, 2022.
Article in English | MEDLINE | ID: mdl-35786602

ABSTRACT

Some population pharmacokinetic models for amiodarone (AMD) did not incorporate N-desethylamiodarone (DEA) concentration. Glucocorticoids activate CYP3A4 activity, metabolizing AMD. In contrast, CYP3A4 activity may decrease under inflammation conditions. However, direct evidence for the role of glucocorticoid or inflammation on the pharmacokinetics of AMD and DEA is lacking. The pilot study aimed to address this gap using a population pharmacokinetic analysis of AMD and DEA. A retrospective cohort observational study in adult patients who underwent AMD treatment with trough concentration measurement was conducted at Tokyo Women's Medical University, Medical Center East from June 2015 to March 2019. Both structural models of AMD and DEA applied 1-compartment models, which included significant covariates using a stepwise forward selection and backward elimination method. The eligible 81 patients (C-reactive protein level: 0.26 [interquartile range; 0.09-1.92] mg/dL) had a total of 408 trough concentrations for both AMD and DEA. The median trough concentrations were 0.49 [0.31-0.81] µg/mL for AMD and 0.43 [0.28-0.71] µg/mL for DEA during a median follow-up period of 446 [147-1059] d. Three patients received low-dose oral glucocorticoid. The final model identified that AMD clearance was 7.9 L/h, and the apparent DEA clearance was 10.3 L/h. Co-administered glucocorticoids lowered apparent DEA clearance by 35%. These results indicate that co-administered glucocorticoids may increase DEA concentrations in patients without severe inflammation.


Subject(s)
Amiodarone , Glucocorticoids , Adult , Amiodarone/analogs & derivatives , Anti-Arrhythmia Agents , Cytochrome P-450 CYP3A , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Pilot Projects , Retrospective Studies
3.
Br J Pharmacol ; 179(13): 3382-3402, 2022 07.
Article in English | MEDLINE | ID: mdl-35106755

ABSTRACT

BACKGROUND AND PURPOSE: The aim of the present study was to study the antiarrhythmic effects and cellular mechanisms of desethylamiodarone (DEA), the main metabolite of amiodarone (AMIO), following acute and chronic 4-week oral treatments (25-50 mg·kg-1 ·day-1 ). EXPERIMENTAL APPROACH: The antiarrhythmic effects of acute iv. (10 mg·kg-1 ) and chronic oral (4 weeks, 25 mg·kg-1 ·day-1 ) administration of DEA were assessed in carbachol and tachypacing-induced dog atrial fibrillation models. Action potentials were recorded from atrial and right ventricular tissue following acute (10 µM) and chronic (p.o. 4 weeks, 50 mg·kg-1 ·day-1 ) DEA application using the conventional microelectrode technique. Ionic currents were measured by the whole cell configuration of the patch clamp technique in isolated left ventricular myocytes. Pharmacokinetic studies were performed following a single intravenous dose (25 mg·kg-1 ) of AMIO and DEA intravenously and orally. In chronic (91-day) toxicological investigations, DEA and AMIO were administered in the oral dose of 25 mg·kg-1 ·day-1 ). KEY RESULTS: DEA exerted marked antiarrhythmic effects in both canine atrial fibrillation models. Both acute and chronic DEA administration prolonged action potential duration in atrial and ventricular muscle without any changes detected in Purkinje fibres. DEA decreased the amplitude of several outward potassium currents such as IKr , IKs , IK1 , Ito , and IKACh , while the ICaL and late INa inward currents were also significantly depressed. Better drug bioavailability and higher volume of distribution for DEA were observed compared to AMIO. No neutropenia and less severe pulmonary fibrosis was found following DEA compared to that of AMIO administration. CONCLUSION AND IMPLICATIONS: Chronic DEA treatment in animal experiments has marked antiarrhythmic and electrophysiological effects with better pharmacokinetics and lower toxicity than its parent compound. These results suggest that the active metabolite, DEA, should be considered for clinical trials as a possible new, more favourable option for the treatment of cardiac arrhythmias including atrial fibrillation.


Subject(s)
Amiodarone , Atrial Fibrillation , Action Potentials , Amiodarone/analogs & derivatives , Amiodarone/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Dogs , Heart Atria , Myocytes, Cardiac
4.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35163464

ABSTRACT

Novel compounds significantly interfering with the mitochondrial energy production may have therapeutic value in triple-negative breast cancer (TNBC). This criterion is clearly fulfilled by desethylamiodarone (DEA), which is a major metabolite of amiodarone, a widely used antiarrhythmic drug, since the DEA previously demonstrated anti-neoplastic, anti-metastasizing, and direct mitochondrial effects in B16F10 melanoma cells. Additionally, the more than fifty years of clinical experience with amiodarone should answer most of the safety concerns about DEA. Accordingly, in the present study, we investigated DEA's potential in TNBC by using a TN and a hormone receptor positive (HR+) BC cell line. DEA reduced the viability, colony formation, and invasive growth of the 4T1 cell line and led to a higher extent of the MCF-7 cell line. It lowered mitochondrial transmembrane potential and induced mitochondrial fragmentation. On the other hand, DEA failed to significantly affect various parameters of the cellular energy metabolism as determined by a Seahorse live cell respirometer. Cyclooxygenase 2 (COX-2), which was upregulated by DEA in the TNBC cell line only, accounted for most of 4T1's DEA resistance, which was counteracted by the selective COX-2 inhibitor celecoxib. All these data indicate that DEA may have potentiality in the therapy of TNBC.


Subject(s)
Amiodarone/analogs & derivatives , Antineoplastic Agents/pharmacology , Celecoxib/pharmacology , Cyclooxygenase 2/metabolism , Mitochondria/metabolism , Triple Negative Breast Neoplasms/metabolism , Amiodarone/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Triple Negative Breast Neoplasms/drug therapy , Up-Regulation/drug effects
5.
Anal Bioanal Chem ; 413(28): 6917-6927, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34595558

ABSTRACT

Quantitative measure of a drug and its associated metabolite(s) with single-cell resolution is often limited by sampling throughput or other compromises that limit broad use. Here, we demonstrate the use of single-cell printing-liquid vortex capture-mass spectrometry (SCP-LVC-MS) to quantitatively measure the intracellular concentrations of amiodarone (AMIO) and its metabolite, N-desethylamiodarone (NDEA), from thousands of single cells across several AMIO incubation concentrations ranging from 0 to 10 µM. Concentrations obtained by SCP-LVC-MS were validated through comparison with average assays and traditional measurement of cells in bulk. Average of SCP-LVC-MS measurements and aggregate vial collection assay the concentrations differed by < 5%. Both AMIO and NDEA had clear log-normal distributions with similar standard deviation of concentrations in the cell population. The mean of both AMIO and NDEA intracellular concentrations were positively correlated with AMIO incubation concentration, increasing from 0.026 to 0.520 and 0.0055 to 0.048 mM for AMIO and NDEA, respectively. The standard deviation of AMIO and NDEA log-normal distribution fits were relatively similar in value across incubation concentrations, 0.15-0.19 log10 (mM), and exhibited a linear trend with respect to each other. The single cell-resolved conversion ratio of AMIO to NDEA increased with decreasing incubation concentration, 7 ± 2%, 18 ± 3%, and 20 ± 7% for 10.0, 1.0, and 0.1 µM AMIO incubation concentrations, respectively. Association with simultaneously measured lipids had several ions with statistically significant difference in intensity but no clear correlations with AMIO intracellular content was observed.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/analysis , Mass Spectrometry/methods , Single-Cell Analysis/methods , Vasodilator Agents/analysis , Hep G2 Cells , Humans , Reproducibility of Results
6.
Drug Metab Pharmacokinet ; 38: 100390, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33836300

ABSTRACT

The combination of antiarrhythmic agents, amiodarone or dronedarone, with the anticoagulant rivaroxaban is used clinically in the management of atrial fibrillation for rhythm control and secondary stroke prevention respectively. Renal drug-drug interactions (DDIs) between amiodarone or dronedarone and rivaroxaban were previously ascribed to inhibition of rivaroxaban secretion by P-glycoprotein at the apical membrane of renal proximal tubular epithelial cells. Benzbromarone, a known inhibitor of organic anion transporter 3 (OAT3), shares a benzofuran scaffold with amiodarone and dronedarone. However, inhibitory activity of amiodarone and dronedarone against OAT3 remains arcane. Here, we conducted in vitro transporter inhibition assays in OAT3-transfected HEK293 cells which revealed amiodarone, dronedarone and their respective major pharmacologically-active metabolites N-desethylamiodarone and N-desbutyldronedarone possess inhibitory activity against OAT3, with corrected Ki values of 0.042, 0.019, 0.028 and 0.0046 µM respectively. Protein binding effects and probe substrate dependency were accounted for in our assays. Static modelling predicted 1.29-, 1.01-, 1.29- and 1.16-fold increase in rivaroxaban exposure, culminating in a predicted 1.29-, 1.01-, 1.28- and 1.15-fold increase in major bleeding risk respectively, suggesting potential OAT3-mediated DDI between amiodarone and rivaroxaban. Future work involving physiologically-based pharmacokinetic modelling is crucial in holistically predicting the complex DDIs between the benzofuran antiarrhythmic agents and rivaroxaban.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzofurans/pharmacology , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amiodarone/analogs & derivatives , Amiodarone/pharmacology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Cell Line , Dronedarone/pharmacology , Drug Interactions/physiology , HEK293 Cells , Humans , Rivaroxaban/pharmacology
7.
Anal Chem ; 92(24): 16005-16015, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33280372

ABSTRACT

The analysis of large numbers of cells from a population results in information that does not reflect differences in cell phenotypes. Individual variations in cellular drug uptake, metabolism, and response to drug treatment may have profound effects on cellular survival and lead to the development of certain disease states, drug persistence, and resistance. Herein, we present a method that combines live cell confocal microscopy imaging with high-resolution mass spectrometry to achieve absolute cell quantification of the drug amiodarone (AMIO) and its major metabolite, N-desethylamiodarone (NDEA), in single liver cells (HepG2 and HepaRG cells). The method uses a prototype system that integrates a confocal microscope with an XYZ stage robot to image and automatically sample selected cells from a sample compartment, which is kept under growth conditions, with nanospray tips. Besides obtaining the distributions of AMIO and NDEA cell concentrations across a population of individual cells, as well as variabilities in drug metabolism, the effect of these on phospholipidosis and cell morphology was studied. The method was suited to identify subpopulations of cells that metabolized less drug and to correlate cell drug concentrations with cell phospholipid content, cell volume, sphericity, and other cell phenotypic features. Using principal component analysis (PCA), the treated cells could be clearly distinguished from vehicle control cells (0 µM AMIO) and HepaRG cells from HepG2 cells. The potential of using multidimensional and multimodal information collected from single cells to build predictive models for cell classification is demonstrated.


Subject(s)
Amiodarone/adverse effects , Amiodarone/metabolism , Lysosomal Storage Diseases/diagnostic imaging , Lysosomal Storage Diseases/pathology , Mass Spectrometry , Microscopy, Confocal/methods , Single-Cell Analysis/methods , Amiodarone/analogs & derivatives , Hep G2 Cells , Humans , Lysosomal Storage Diseases/chemically induced
8.
Int J Mol Sci ; 21(19)2020 Oct 05.
Article in English | MEDLINE | ID: mdl-33027919

ABSTRACT

Previously, we showed that desethylamiodarone (DEA), a major metabolite of the widely used antiarrhythmic drug amiodarone, has direct mitochondrial effects. We hypothesized that these effects account for its observed cytotoxic properties and ability to limit in vivo metastasis. Accordingly, we examined DEA's rapid (3-12 h) cytotoxicity and its early (3-6 h) effects on various mitochondrial processes in B16F10 melanoma cells. DEA did not affect cellular oxygen radical formation, as determined using two fluorescent dyes. However, it did decrease the mitochondrial transmembrane potential, as assessed by JC-1 dye and fluorescence microscopy. It also induced mitochondrial fragmentation, as visualized by confocal fluorescence microscopy. DEA decreased maximal respiration, ATP production, coupling efficiency, glycolysis, and non-mitochondrial oxygen consumption measured by a Seahorse cellular energy metabolism analyzer. In addition, it induced a cyclosporine A-independent mitochondrial permeability transition, as determined by Co2+-mediated calcein fluorescence quenching measured using a high-content imaging system. DEA also caused outer mitochondrial membrane permeabilization, as assessed by the immunoblot analysis of cytochrome C, apoptosis inducing factor, Akt, phospho-Akt, Bad, and phospho-Bad. All of these data supported our initial hypothesis.


Subject(s)
Amiodarone/analogs & derivatives , Cell Proliferation/drug effects , Melanoma, Experimental/drug therapy , Mitochondria/genetics , Amiodarone/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor , Cytochromes c/genetics , Cytostatic Agents/pharmacology , Energy Metabolism/drug effects , Humans , Lung/metabolism , Lung/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Oxygen Consumption/drug effects , Permeability/drug effects , Reactive Oxygen Species/metabolism
9.
PLoS One ; 15(9): e0239088, 2020.
Article in English | MEDLINE | ID: mdl-32977329

ABSTRACT

Previously, we demonstrated the in vitro anti-tumor effects of desethylamiodarone (DEA) in bladder and cervix cancer cell lines. In the present study, we intended to establish its potentiality in B16-F10 metastatic melanoma cells in vitro and in vivo. We assessed cell proliferation, apoptosis and cell cycle by using sulforhodamine B assay, Muse™ Annexin V & Dead Cell and Muse® Cell Cycle assays, respectively. We determined colony formation after crystal violet staining. For studying mechanistic aspects, immunoblotting analysis was performed. We used a C57BL/6 experimental lung metastasis model for demonstrating in vivo anti-metastatic potential of DEA. DEA inhibited in vitro proliferation and colony formation, and in vivo lung metastasizing properties of B16-F10 cells. It arrested the cells in G0/G1 phase of their cycle likely via p21 in a p53-dependent fashion, and induced caspase mediated apoptosis likely via inversely regulating Bcl-2 and Bax levels, and reducing Akt and ERK1/2 activation. In this study, we provided in vitro and in vivo experimental evidences for DEA's potentiality in the therapy of metastatic melanomas. Since DEA is the major metabolite of amiodarone, a worldwide used antiarrhythmic drug, safety concerns could be resolved more easily for it than for a novel pharmacological agent.


Subject(s)
Amiodarone/analogs & derivatives , Antineoplastic Agents/therapeutic use , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Amiodarone/therapeutic use , Animals , Anti-Arrhythmia Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Skin Neoplasms/pathology
11.
Can J Physiol Pharmacol ; 96(10): 1004-1011, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29847733

ABSTRACT

Previously, we found that desethylamiodarone (DEA) may have therapeutic potentiality in bladder cancer. In this study, we determined its effects on human cervical cancer cells (HeLa). Cell viability was evaluated by Muse Cell Count & Viability Assay; cell apoptosis was detected by Muse Annexin V & Dead Cell Assay. Cell cycle was flow cytometrically determined by Muse Cell Cycle Kit and the morphological changes of the cells were observed under a fluorescence microscope after Hoechst 33342 staining. The changes in the expression levels of apoptosis-related proteins in the HeLa cells were assessed by immunoblot. Our results showed that DEA significantly inhibited the proliferation and viability of HeLa cells and induced apoptosis in vitro in dose-dependent and also in cell cycle-dependent manner because DEA induced G0/G1 phase arrest in the HeLa cell line. We found that DEA treatment downregulated the expression of phospho-Akt and phospho-Bad. In addition, DEA could downregulate expression of Bcl-2, upregulate Bax, and induce cytochrome c release. Our results indicate that DEA might have significance as an anti-tumor agent against human cervical cancer.


Subject(s)
Amiodarone/analogs & derivatives , Apoptosis/drug effects , Uterine Cervical Neoplasms/pathology , Amiodarone/metabolism , Amiodarone/pharmacology , Cell Cycle Checkpoints/drug effects , Female , HeLa Cells , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
13.
Exp Parasitol ; 189: 8-15, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29684665

ABSTRACT

Chagas disease is a neglected tropical affection caused by the protozoan parasite Trypanosoma cruzi. There is no current effective treatment since the only two available drugs have a limited efficacy and produce side effects. Thus, investigation efforts have been directed to the identification of new drug leads. In this context, Ca2+ regulating mechanisms have been postulated as targets for antiparasitic compounds, since they present paramount differences when compared to host cells. Amiodarone is an antiarrhythmic with demonstrated trypanocidal activity acting through the disruption of the parasite intracellular Ca2+ homeostasis. We now report the effect of a benzofuran derivative based on the structure of amiodarone on T. cruzi. This derivative was able to inhibit the growth of epimastigotes in culture and of amastigotes inside infected cells, the clinically relevant phase. We also show that this compound, similarly to amiodarone, disrupts Ca2+ homeostasis in T. cruzi epimastigotes, via two organelles involved in the intracellular Ca2+ regulation and the bioenergetics of the parasite. We demonstrate that the benzofuran derivative was able to totally collapse the membrane potential of the unique giant mitochondrion of the parasite and simultaneously produced the alkalinization of the acidocalcisomes. Both effects are evidenced by a large increase in the intracellular Ca2+ concentration of T. cruzi.


Subject(s)
Benzofurans/pharmacology , Chagas Disease/drug therapy , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Amiodarone/analogs & derivatives , Amiodarone/chemistry , Amiodarone/pharmacology , Animals , Benzofurans/chemistry , Benzofurans/therapeutic use , Calcium/metabolism , Chlorocebus aethiops , Dose-Response Relationship, Drug , Dronedarone , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Oxidoreductases/metabolism , Trypanocidal Agents/chemistry , Trypanocidal Agents/therapeutic use , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/metabolism , Vero Cells
14.
Environ Mol Mutagen ; 59(4): 278-289, 2018 05.
Article in English | MEDLINE | ID: mdl-29399883

ABSTRACT

Dronedarone, an antiarrhythmic drug, has been marketed as an alternative to amiodarone. The use of dronedarone has been associated with severe liver injury; however, the mechanisms remain unclear. In this study, the possible mechanisms of dronedarone induced liver toxicity were characterized in HepG2 cells. Dronedarone decreased cells viability and induced apoptosis and DNA damage in a concentration- and time-dependent manner. Pretreatment of the HepG2 cells with apoptosis inhibitors (caspase-3, -8, and -9) or the necrosis inhibitor (Necrox-5), partially, but significantly, reduced the release of lactate dehydrogenase. Dronedarone caused the release of cytochrome c from mitochondria to cytosol, a prominent feature of apoptosis. In addition, the activation of caspase-2 was involved in dronedarone induced DNA damage and the activation of JNK and p38 signaling pathways. Inhibition of JNK and p38 by specific inhibitors attenuated dronedarone-induced cell death, apoptosis, and DNA damage. Additionally, suppression of caspase-2 decreased the activities of JNK and p38. Dronedarone triggered DNA damage was regulated by downregulation of topoisomerase IIα at both transcriptional and post-transcriptional levels. Taken together, our data show that DNA damage, apoptosis, and the activation of JNK and p38 contribute to dronedarone-induced cytotoxicity. Environ. Mol. Mutagen. 59:278-289, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Amiodarone/analogs & derivatives , Apoptosis , Cell Survival/drug effects , DNA Damage , Hepatocytes/drug effects , Mitogen-Activated Protein Kinases/metabolism , Amiodarone/toxicity , Down-Regulation , Dronedarone , Hep G2 Cells , Hepatocytes/enzymology , Humans , Signal Transduction
15.
Biochemistry ; 57(5): 805-816, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29200287

ABSTRACT

Heterotropic interactions between atorvastatin (ARVS) and dronedarone (DND) have been deciphered using global analysis of the results of binding and turnover experiments for pure drugs and their mixtures. The in vivo presence of atorvastatin lactone (ARVL) was explicitly taken into account by using pure ARVL in analogous experiments. Both ARVL and ARVS inhibit DND binding and metabolism, while a significantly higher affinity of CYP3A4 for ARVL makes the latter the main modulator of activity (effector) in this system. Molecular dynamics simulations reveal significantly different modes of interactions of DND and ARVL with the substrate binding pocket and with a peripheral allosteric site. Interactions of both substrates with residues F213 and F219 at the allosteric site play a critical role in the communication of conformational changes induced by effector binding to productive binding of the substrate at the catalytic site.


Subject(s)
Amiodarone/analogs & derivatives , Atorvastatin/metabolism , Cytochrome P-450 CYP3A/metabolism , Allosteric Regulation , Allosteric Site , Amiodarone/metabolism , Animals , Binding Sites , Dose-Response Relationship, Drug , Dronedarone , Drug Interactions , Humans , Kinetics , Models, Molecular , Molecular Dynamics Simulation , NADPH-Ferrihemoprotein Reductase/metabolism , Protein Binding , Protein Conformation , Rats
16.
Clin Drug Investig ; 38(1): 39-48, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29080209

ABSTRACT

OBJECTIVE: This retrospective cohort study was performed to examine the association between serum amiodarone (AMD) and N-desethylamiodarone (DEA) concentrations and the development of thyroid dysfunction. METHODS: Patients treated with AMD from January 2012 to April 2016 were identified from the computerized hospital information system database at the National Cerebral and Cardiovascular Center. Only patients whose serum AMD and DEA concentrations had been determined at least once were included in the study. RESULTS: A total of 377 patients were enrolled. Consequently, 54 (14.3%) and 60 (15.9%) patients who developed AMD-induced thyrotoxicosis and hypothyroidism were included. The mean DEA/AMD ratio during the pre-index period in the thyrotoxicosis group (0.86 ± 0.24) was significantly higher than in the hypothyroidism (0.68 ± 0.27) and euthyroidism (0.78 ± 0.30; p < 0.0001) groups. In addition, the mean DEA/AMD ratio during the post-index period in the thyrotoxicosis group (1.05 ± 0.40) was significantly higher than in the hypothyroidism (0.81 ± 0.24) and euthyroidism (0.88 ± 0.22; p < 0.0001) groups. A persistently higher DEA/AMD ratio was observed throughout the study period in the thyrotoxicosis group. In addition, good correlations between the DEA/AMD ratio and the levels of free thyroxine, free triiodothyronine levels, and log (thyroid-stimulating hormone) were observed in the thyrotoxicosis and euthyroidism groups. CONCLUSION: Patients with AMD-induced thyrotoxicosis had an increased DEA/AMD ratio and patients with AMD-induced hypothyroidism had a decreased DEA/AMD ratio before the development of thyroid dysfunction. The DEA/AMD ratio may be a predictive marker for AMD-induced thyroid dysfunction.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/blood , Hypothyroidism/epidemiology , Thyrotoxicosis/epidemiology , Adult , Aged , Aged, 80 and over , Amiodarone/adverse effects , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/blood , Female , Humans , Hypothyroidism/chemically induced , Male , Middle Aged , Retrospective Studies , Thyrotoxicosis/chemically induced , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood
17.
Eur J Pharm Sci ; 111: 13-19, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28942006

ABSTRACT

Dronedarone is a CYP2D6 inhibitor; therefore, it is prudent to exercise caution when concurrently administering CYP2D6-metabolized ß-blockers because of a lack of published data on potential drug interactions. The aim of this study was to investigate the effect of dronedarone on the pharmacokinetics of orally administered carvedilol in rats. Twenty male Sprague-Dawley rats were randomly divided into two groups and 10mg/kg carvedilol was administered to the rat with or without dronedarone pretreatment in a parallel design. Blood samples were collected before and after 0.25, 0.5, 0.75, 1, 2, 4, 6, 8, 12, and 24h of drug administration. The plasma concentration of carvedilol was determined using LC-MS/MS. The systemic exposure to carvedilol was significantly increased and elimination of carvedilol was significantly decreased in the dronedarone-pretreated rats than in the vehicle-pretreated rats. The one-compartment model with first-order absorption and elimination was sufficient to explain the pharmacokinetic characters after single oral administration of carvedilol to both vehicle-pretreated and dronedarone-pretreated rats. This study suggests that dronedarone inhibits CYP2D6-mediated carvedilol metabolism, and dose adjustment is needed in carvedilol and dronedarone combination therapy. Further studies are needed to clarify the effect of dronedarone on carvedilol and CYP2D6 substrates in clinical use.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Amiodarone/analogs & derivatives , Carbazoles/pharmacokinetics , Cytochrome P-450 CYP2D6 Inhibitors/pharmacology , Models, Biological , Propanolamines/pharmacokinetics , Administration, Oral , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/blood , Amiodarone/administration & dosage , Amiodarone/pharmacology , Animals , Carbazoles/administration & dosage , Carbazoles/blood , Carvedilol , Cytochrome P-450 CYP2D6 Inhibitors/administration & dosage , Dronedarone , Drug Interactions , Male , Propanolamines/administration & dosage , Propanolamines/blood , Rats, Sprague-Dawley , Substrate Specificity
18.
PLoS One ; 12(12): e0189470, 2017.
Article in English | MEDLINE | ID: mdl-29220397

ABSTRACT

Bladder cancer (BC) is a common malignancy of the urinary tract that has a higher frequency in men than in women. Cytostatic resistance and metastasis formation are significant risk factors in BC therapy; therefore, there is great interest in overcoming drug resistance and in initiating research for novel chemotherapeutic approaches. Here, we suggest that desethylamiodarone (DEA)-a metabolite of amiodarone-may have cytostatic potential. DEA activates the collapse of mitochondrial membrane potential (detected by JC-1 fluorescence), and induces cell death in T24 human transitional-cell bladder carcinoma cell line at physiologically achievable concentrations. DEA induces cell cycle arrest in the G0/G1 phase, which may contribute to the inhibition of cell proliferation, and shifts the Bax/Bcl-2 ratio to initiate apoptosis, induce AIF nuclear translocation, and activate PARP-1 cleavage and caspase-3 activation. The major cytoprotective kinases-ERK and Akt-are inhibited by DEA, which may contribute to its cell death-inducing effects. DEA also inhibits the expression of B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1) and reduces colony formation of T24 bladder carcinoma cells, indicating its possible inhibitory effect on metastatic potential. These data show that DEA is a novel anti-cancer candidate of multiple cell death-inducing effects and metastatic potential. Our findings recommend further evaluation of its effects in clinical studies.


Subject(s)
Amiodarone/analogs & derivatives , Apoptosis/drug effects , Urinary Bladder Neoplasms/pathology , Amiodarone/pharmacology , Cell Line, Tumor , Humans , Membrane Potential, Mitochondrial/drug effects
19.
PLoS One ; 12(11): e0188442, 2017.
Article in English | MEDLINE | ID: mdl-29161309

ABSTRACT

BACKGROUND AND AIMS: Left ventricular hypertrophy (LVH) in hypertension is associated with a greater risk of sustained supraventricular/atrial arrhythmias. Dronedarone is an antiarrhythmic agent that was recently approved for the treatment of atrial fibrillation. However, its effect on early regression of LVH has not been reported. We tested the hypothesis that short-term administration of dronedarone induces early regression of LVH in spontaneously hypertensive rats (SHRs). METHODS: Ten-month-old male SHRs were randomly assigned to an intervention group (SHR-D), where animals received dronedarone treatment (100 mg/kg) for a period of 14 days, or to a control group (SHR) where rats were given vehicle. A third group with normotensive control rats (WKY) was also added. At the end of the treatment with dronedarone we studied the cardiac anatomy and function in all the rats using transthoracic echocardiogram, cardiac metabolism using the PET/CT study (2-deoxy-2[18F]fluoro-D-glucose) and cardiac structure by histological analysis of myocyte size and collagen content. RESULTS: The hypertensive vehicle treated SHR rats developed the classic cardiac pattern of hypertensive cardiomyopathy as expected for the experimental model, with increases in left ventricular wall thickness, a metabolic shift towards an increase in glucose use and increases in myocyte and collagen content. However, the SHR-D rats showed statistically significant lower values in comparison to SHR group for septal wall thickness, posterior wall thickness, ventricular mass, glucose myocardial uptake, size of left ventricular cardiomyocytes and collagen content. All these values obtained in SHR-D rats were similar to the values measured in the normotensive WKY control group. CONCLUSION: The results suggest by three alternative and complementary ways (analysis of anatomy and cardiac function, metabolism and histological structure) that dronedarone has the potential to reverse the LVH induced by arterial hypertension in the SHR model of compensated ventricular hypertrophy.


Subject(s)
Amiodarone/analogs & derivatives , Heart Diseases/drug therapy , Hypertension/drug therapy , Hypertrophy, Left Ventricular/drug therapy , Amiodarone/administration & dosage , Animals , Atrial Fibrillation/diagnostic imaging , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Atrial Remodeling/drug effects , Dronedarone , Heart Diseases/diagnostic imaging , Heart Diseases/physiopathology , Humans , Hypertension/diagnostic imaging , Hypertension/physiopathology , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/physiopathology , Positron Emission Tomography Computed Tomography , Rats , Rats, Inbred SHR/physiology
20.
Atherosclerosis ; 266: 81-86, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28992468

ABSTRACT

BACKGROUND AND AIMS: Dronedarone reduced the rate of stroke and transient ischemic attack in patients with paroxysmal atrial fibrillation (AF) in the ATHENA trial. This cannot be explained by its antiarrhythmic effect alone and may involve alternative mechanisms. This study aimed to investigate any direct effect of dronedarone on blood thrombogenicity, independent of its antiarrhythmic effects. METHODS: Blood samples from patients with cardiovascular disease (n = 30) taking no anticoagulant or antiplatelet medication except aspirin were incubated with dronedarone's active metabolite (SR35021A) at 66 ng/ml (am-L) and 119 ng/ml (am-H), i.e., minimum and maximum mean Cmax reported after repeated 400 mg BID dosing. A third aliquot of blood was incubated with vehicle (Control). Antithrombotic effects of dronedarone were assessed using Coagulation Time (CT), Clot Formation Rate (CFR) and Maximum Clot Firmness (MCF) in ThromboElastoMetry and whole blood platelet aggregation in response to ADP, collagen and TRAP. RESULTS: Compared to Control, mean CT was prolonged by am-L and am-H (164 ± 25 s vs. 180 ± 22 and 182 ± 32 s, respectively, p<0.01 for both). Small but statistically significant reductions were observed in CFR (am-L and am-H) and MCF (am-H). Platelet aggregation induced by ADP and TRAP was also reduced (p<0.05 for both) by am-H. CONCLUSIONS: Dronedarone exerts direct anticoagulant and antiplatelet effects on human blood in vitro that are independent of its antiarrhythmic actions. This suggests the reductions in ischemic events reported with dronedarone may not be due to amelioration of AF itself. Additional clinical studies are required to further improve understanding of the mechanisms involved.


Subject(s)
Amiodarone/analogs & derivatives , Anti-Arrhythmia Agents/pharmacology , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Aged , Amiodarone/pharmacology , Blood Coagulation Tests , Cardiovascular Diseases/blood , Dronedarone , Female , Humans , Male , Middle Aged , Platelet Function Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...