Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 760
Filter
Add more filters

Publication year range
1.
Int J Clin Pharmacol Ther ; 62(9): 402-411, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39045733

ABSTRACT

OBJECTIVE: High plasma levels of mono-N-desethylamiodarone (MDEA), an active amiodarone metabolite, may be associated with tissue toxicity in heart failure (patients with heart rhythm disturbances); therefore, a tool that can identify patients for whom therapeutic drug monitoring (TDM) of MDEA is required. This multicenter study aimed to develop a decision tree (DT) model that can identify patients with heart rhythm disturbances at high MDEA concentrations. MATERIALS AND METHODS: A multicenter retrospective cohort study was conducted, including 157 adult patients with heart failure who received oral amiodarone treatment. A χ2 automatic interaction-detection algorithm was used to construct a DT model. In the DT analysis, the dependent variable was set as an MDEA trough plasma concentration of ≥ 0.6 µg/mL during the steady-state period. Explanatory variables were selected as factors with p < 0.05 in multivariate logistic regression analysis. RESULTS: The adjusted odds ratios for the daily dose of amiodarone and body mass index were 1.01 (95% coefficient interval: 1.008 - 1.021, p < 0.001) and 0.91 (95% confidence interval: 0.834 - 0.988, p = 0.025), respectively. For DT analysis, the risk of reaching plasma MDEA concentrations ≥ 0.6 µg/mL was relatively high, combined with a daily dose of amiodarone > 100 mg and body mass index ≤ 22.3 kg/m2 at 69.0% (20/29), and its trend was also detected in the sensitivity analysis. CONCLUSION: Patients taking a daily amiodarone dose > 100 mg and with a body mass index ≤ 22.3 kg/m2 warrant TDM implementation for MDEA to minimize the risk of MDEA-induced tissue toxicity.


Subject(s)
Amiodarone , Anti-Arrhythmia Agents , Decision Trees , Drug Monitoring , Humans , Amiodarone/adverse effects , Amiodarone/administration & dosage , Amiodarone/pharmacokinetics , Amiodarone/analogs & derivatives , Retrospective Studies , Male , Female , Aged , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/blood , Middle Aged , Drug Monitoring/methods , Heart Failure/drug therapy , Aged, 80 and over
2.
Int J Mol Sci ; 25(18)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39337269

ABSTRACT

Previously, we have demonstrated that amiodarone (AM), a widely used antiarrhythmic drug, and its major metabolite desethylamiodarone (DEA) both affect several mitochondrial processes in isolated heart and liver mitochondria. Also, we have established DEA's antitumor properties in various cancer cell lines and in a rodent metastasis model. In the present study, we compared AM's and DEA's mitochondrial and antineoplastic effects in a human triple-negative breast cancer (TNBC) cell line. Both compounds reduced viability in monolayer and sphere cultures and the invasive growth of the MDA-MB-231 TNBC line by inducing apoptosis. They lowered mitochondrial trans-membrane potential, increased Ca2+ influx, induced mitochondrial permeability transition, and promoted mitochondrial fragmentation. In accordance with their mitochondrial effects, both substances massively decreased overall, and even to a greater extent, mitochondrial ATP production decreased, as determined using a Seahorse live cell respirometer. In all these effects, DEA was more effective than AM, indicating that DEA may have higher potential in the therapy of TNBC than its parent compound.


Subject(s)
Amiodarone , Antineoplastic Agents , Apoptosis , Mitochondria , Triple Negative Breast Neoplasms , Amiodarone/pharmacology , Amiodarone/analogs & derivatives , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Female , Membrane Potential, Mitochondrial/drug effects , Cell Survival/drug effects , Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Proliferation/drug effects
3.
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
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.
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
7.
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
8.
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
9.
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
10.
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
11.
J Cell Mol Med ; 21(10): 2514-2523, 2017 10.
Article in English | MEDLINE | ID: mdl-28425222

ABSTRACT

Drug-induced ion channel trafficking disturbance can cause cardiac arrhythmias. The subcellular level at which drugs interfere in trafficking pathways is largely unknown. KIR 2.1 inward rectifier channels, largely responsible for the cardiac inward rectifier current (IK1 ), are degraded in lysosomes. Amiodarone and dronedarone are class III antiarrhythmics. Chronic use of amiodarone, and to a lesser extent dronedarone, causes serious adverse effects to several organs and tissue types, including the heart. Both drugs have been described to interfere in the late-endosome/lysosome system. Here we defined the potential interference in KIR 2.1 backward trafficking by amiodarone and dronedarone. Both drugs inhibited IK1 in isolated rabbit ventricular cardiomyocytes at supraclinical doses only. In HK-KWGF cells, both drugs dose- and time-dependently increased KIR 2.1 expression (2.0 ± 0.2-fold with amiodarone: 10 µM, 24 hrs; 2.3 ± 0.3-fold with dronedarone: 5 µM, 24 hrs) and late-endosomal/lysosomal KIR 2.1 accumulation. Increased KIR 2.1 expression level was also observed in the presence of Nav 1.5 co-expression. Augmented KIR 2.1 protein levels and intracellular accumulation were also observed in COS-7, END-2, MES-1 and EPI-7 cells. Both drugs had no effect on Kv 11.1 ion channel protein expression levels. Finally, amiodarone (73.3 ± 10.3% P < 0.05 at -120 mV, 5 µM) enhanced IKIR2.1 upon 24-hrs treatment, whereas dronedarone tended to increase IKIR2.1 and it did not reach significance (43.8 ± 5.5%, P = 0.26 at -120 mV; 2 µM). We conclude that chronic amiodarone, and potentially also dronedarone, treatment can result in enhanced IK1 by inhibiting KIR 2.1 degradation.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/pharmacology , Ion Channel Gating/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , COS Cells , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Dronedarone , HEK293 Cells , Humans , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/genetics , Rabbits
12.
Ann Pharmacother ; 51(3): 245-252, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27758968

ABSTRACT

OBJECTIVE: To review the limitations of current antiarrhythmic drugs in atrial fibrillation (AF) and discuss the rationale and clinical trials supporting the use of ranolazine in AF. DATA SOURCES: MEDLINE was searched from 1980 to September 2016 using the terms ranolazine, atrial fibrillation, coronary artery bypass grafting, and valve surgery. STUDY SELECTION AND DATA EXTRACTION: English-language studies and reviews assessing antiarrhythmic drugs, including ranolazine, were incorporated. DATA SYNTHESIS: The use of ranolazine monotherapy has been evaluated in 2 clinical trials. In the RAFFAELLO trial, higher doses of ranolazine showed a trend toward lower AF recurrence versus placebo ( P = 0.053), but further evidence is needed to support its use as a sole therapeutic agent. Ranolazine has shown utility in a limited number of studies as an adjunctive agent, which is critical for those in whom standard therapy is inadequate or the adverse event profile precludes optimized standard therapy. In the HARMONY trial, ranolazine 750 mg and dronedarone 225 mg twice daily reduced the AF burden by 59.1% from baseline ( P = 0.008 vs placebo). In a trial by Koskinas and colleagues, patients receiving ranolazine 1500 mg once and intravenous amiodarone had a higher conversion rate than those receiving amiodarone alone ( P = 0.024). There are also promising studies for the prevention and treatment of post-cardiothoracic surgery AF, which require further investigation. CONCLUSIONS: Ranolazine's pharmacological properties and available evidence suggest potential for its use in AF.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Ranolazine/therapeutic use , Amiodarone/administration & dosage , Amiodarone/adverse effects , Amiodarone/analogs & derivatives , Amiodarone/therapeutic use , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/epidemiology , Cardiac Surgical Procedures/adverse effects , Clinical Trials as Topic , Dronedarone , Drug Therapy, Combination , Humans , Male , Ranolazine/administration & dosage , Ranolazine/adverse effects , Recurrence , Treatment Outcome
13.
Eur J Clin Pharmacol ; 73(3): 289-296, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28083650

ABSTRACT

PURPOSE: We used a retrospective data mining approach to explore the association between serum amiodarone (AMD) and N-desethylamiodarone (DEA) concentrations and thyroid-related hormone levels. METHODS: Laboratory data sets from January 2012 to April 2016 were extracted from the computerized hospital information system database at the National Cerebral and Cardiovascular Center (NCVC). Data sets that contained serum AMD and DEA concentrations and thyroid function tests, including thyroid-stimulating hormone (TSH), free thyroxine (FT4), and free triiodothyronine (FT3), were analyzed. RESULTS: A total of 1831 clinical laboratory data sets from 330 patients were analyzed. Data sets were classified into five groups (euthyroidism, hyperthyroidism, subclinical hyperthyroidism, hypothyroidism, and subclinical hypothyroidism) based on the definition of thyroid function in our hospital. Most abnormal levels of thyroid hormones were observed within the therapeutic range of serum AMD and DEA concentrations. The mean DEA/AMD ratio in the hyperthyroidism group was significantly higher than that in the euthyroidism group (0.95 ± 0.42 vs. 0.87 ± 0.28, p = 0.0209), and the mean DEA/AMD ratio in the hypothyroidism group was significantly lower than that in the euthyroidism group (0.77 ± 0.26 vs. 0.87 ± 0.28, p = 0.0038). The suppressed TSH group (0.98 ± 0.41 vs. 0.87 ± 0.28, p < 0.001) and the elevated FT4 level group (0.90 ± 0.33 vs. 0.84 ± 0.27, p = 0.0037) showed significantly higher DEA/AMD ratios compared with normal level groups. The elevated TSH group showed a significantly lower DEA/AMD ratio compared with the normal group (0.81 ± 0.25 vs. 0.87 ± 0.28, p < 0.0001). CONCLUSIONS: High and low DEA/AMD ratios were associated with AMD-induced hyperthyroidism and hypothyroidism, respectively. The DEA/AMD ratio may be a predictive marker for AMD-induced thyroid dysfunction.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/blood , Hyperthyroidism/chemically induced , Hypothyroidism/chemically induced , Amiodarone/adverse effects , Female , Humans , Male , Retrospective Studies , Thyrotropin/blood , Thyroxine/blood
14.
Pharmacology ; 99(3-4): 128-138, 2017.
Article in English | MEDLINE | ID: mdl-27992867

ABSTRACT

We have previously reported that amiodarone interacts with a novel allosteric site on muscarinic receptors. Amiodarone's most striking effect is to enhance the maximal response elicited by muscarinic agonists at the M1, M3, and M5 receptors. Furthermore, the quaternary analog N-ethylamiodarone (NEA) is inhibitory at these receptors and appears to compete with amiodarone at that allosteric site. In the present studies, we show that dronedarone also modulates Gq-mediated responses at M1 and M3, although in a more discriminating manner. For example, dronedarone markedly enhances pilocarpine-stimulated release of arachidonic acid from CHO cells, via the M3 receptor subtype, but does not affect the acetylcholine-stimulated response. Such probe-dependent effects are diagnostic of an allosteric interaction. In comparison to these effects at M3, dronedarone is strongly inhibitory toward both pilocarpine and acetylcholine at the M1 subtype. The effects of dronedarone are consistent with an interaction at the amiodarone site: dronedarone inhibits the enhancement of acetylcholine's response produced by amiodarone at the M3 subtype; also, NEA reverses the enhancement of pilocarpine's response at M3 produced by either dronedarone or amiodarone. In studies with the M1-selective allosteric agonist 4-[3-(4-butylpiperidin-1-yl)-propyl]-7-fluoro-4H-benzo[1,4]oxazin-3-one (AC-260584), amiodarone enhanced the maximal response observed, whereas dronedarone was inhibitory. On the other hand, benzyl quinolone carboxylic acid, the well-known allosteric ligand that dramatically enhances the potency of acetylcholine at the M1 subtype, had no effect on the response profile of AC-260584. In summary, dronedarone acts at M1 and M3 muscarinic receptors in a manner that complements amiodarone and provides an additional tool with which to investigate this novel allosteric site.


Subject(s)
Amiodarone/analogs & derivatives , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Amiodarone/chemistry , Amiodarone/metabolism , Amiodarone/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Dronedarone
15.
J Mol Cell Cardiol ; 94: 95-106, 2016 05.
Article in English | MEDLINE | ID: mdl-27056421

ABSTRACT

INTRODUCTION: Pharmacological rhythm control of atrial fibrillation (AF) in patients with structural heart disease is limited. Ranolazine in combination with low dose dronedarone remarkably reduced AF-burden in the phase II HARMONY trial. We thus aimed to investigate the possible mechanisms underlying these results. METHODS AND RESULTS: Patch clamp experiments revealed that ranolazine (5µM), low-dose dronedarone (0.3µM), and the combination significantly prolonged action potential duration (APD90) in atrial myocytes from patients in sinus rhythm (prolongation by 23.5±0.1%, 31.7±0.1% and 25.6±0.1% respectively). Most importantly, in atrial myocytes from patients with AF ranolazine alone, but more the combination with dronedarone, also prolonged the typically abbreviated APD90 (prolongation by 21.6±0.1% and 31.9±0.1% respectively). It was clearly observed that neither ranolazine, dronedarone nor the combination significantly changed the APD or contractility and twitch force in ventricular myocytes or trabeculae from patients with heart failure (HF). Interestingly ranolazine, and more so the combination, but not dronedarone alone, caused hyperpolarization of the resting membrane potential in cardiomyocytes from AF. As measured by confocal microscopy (Fluo-3), ranolazine, dronedarone and the combination significantly suppressed diastolic sarcoplasmic reticulum (SR) Ca(2+) leak in myocytes from sinus rhythm (reduction by ranolazine: 89.0±30.7%, dronedarone: 75.6±27.4% and combination: 78.0±27.2%), in myocytes from AF (reduction by ranolazine: 67.6±33.7%, dronedarone: 86.5±31.7% and combination: 81.0±33.3%), as well as in myocytes from HF (reduction by ranolazine: 64.8±26.5% and dronedarone: 65.9±29.3%). CONCLUSIONS: Electrophysiological measurements during exposure to ranolazine alone or in combination with low-dose dronedarone showed APD prolongation, cellular hyperpolarization and reduced SR Ca(2+) leak in human atrial myocytes. The combined inhibitory effects on various currents, in particular Na(+) and K(+) currents, may explain the anti-AF effects observed in the HARMONY trial. Therefore, the combination of ranolazine and dronedarone, but also ranolazine alone, may be promising new treatment options for AF, especially in patients with HF, and merit further clinical investigation.


Subject(s)
Amiodarone/analogs & derivatives , Atrial Function/drug effects , Heart Atria/drug effects , Heart Ventricles/drug effects , Ranolazine/pharmacology , Ventricular Function/drug effects , Aged , Amiodarone/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cardiovascular Agents/pharmacology , Dronedarone , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sarcomeres/drug effects , Sarcomeres/metabolism
16.
Mol Pharmacol ; 89(1): 1-13, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26490246

ABSTRACT

Dronedarone is an antiarrhythmic agent approved in 2009 for the treatment of atrial fibrillation. An in-house preliminary study demonstrated that dronedarone inhibits cytochrome P450 (CYP) 3A4 and 3A5 in a time-dependent manner. This study aimed to investigate the inactivation of CYP450 by dronedarone. We demonstrated for the first time that both dronedarone and its main metabolite N-desbutyl dronedarone (NDBD) inactivate CYP3A4 and CYP3A5 in a time-, concentration-, and NADPH-dependent manner. For the inactivation of CYP3A4, the inactivator concentration at the half-maximum rate of inactivation and inactivation rate constant at an infinite inactivator concentration are 0.87 µM and 0.039 minute(-1), respectively, for dronedarone, and 6.24 µM and 0.099 minute(-1), respectively, for NDBD. For CYP3A5 inactivation, the inactivator concentration at the half-maximum rate of inactivation and inactivation rate constant at an infinite inactivator concentration are 2.19 µM and 0.0056 minute(-1) for dronedarone and 5.45 µM and 0.056 minute(-1) for NDBD. The partition ratios for the inactivation of CYP3A4 and CYP3A5 by dronedarone are 51.1 and 32.2, and the partition ratios for the inactivation of CYP3A4 and CYP3A5 by NDBD are 35.3 and 36.6. Testosterone protected both CYP3A4 and CYP3A5 from inactivation by dronedarone and NDBD. Although the presence of Soret peak confirmed the formation of a quasi-irreversible metabolite-intermediate complex between dronedarone/NDBD and CYP3A4/CYP3A5, partial recovery of enzyme activity by potassium ferricyanide illuminated an alternative irreversible mechanism-based inactivation (MBI). MBI of CYP3A4 and CYP3A5 was further supported by the discovery of glutathione adducts derived from the quinone oxime intermediates of dronedarone and NDBD. In conclusion, dronedarone and NDBD inactivate CYP3A4 and CYP3A5 via unique dual mechanisms of MBI and formation of the metabolite-intermediate complex. Our novel findings contribute new knowledge for future investigation of the underlying mechanisms associated with dronedarone-induced hepatotoxicity and clinical drug-drug interactions.


Subject(s)
Amiodarone/analogs & derivatives , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Amiodarone/chemistry , Amiodarone/metabolism , Amiodarone/pharmacology , Cytochrome P-450 Enzyme Inhibitors/chemistry , Dose-Response Relationship, Drug , Dronedarone , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology
17.
Cell Physiol Biochem ; 38(6): 2311-22, 2016.
Article in English | MEDLINE | ID: mdl-27197836

ABSTRACT

BACKGROUND/AIMS: Amiodarone, a thyroid hormone-like molecule, can induce dyslipidemia and thyroid dysfunction. However, the effects of dronedarone on lipid metabolism and of both dronedarone and amiodarone on thyroid function and lipid metabolism remain unknown. METHODS: Fifty male Sprague-Dawley rats were randomly divided into 5 groups (10 in each group): normal control (NC), amiodarone-treated (AMT), dronedarone-treated (DRT), rats treated with amiodarone combined with polyene phosphatidylcholine (AC), and rats treated with dronedarone combined with polyene phosphatidylcholine (DC). Rats were given amiodarone (120 mg/kg/d), dronedarone (120 mg/kg/d), and polyene phosphatidylcholine (200 mg/kg/d) for 13 weeks. At the end of weeks 4, 8, 12, and 13, plasma-free triiodothyronine (FT3), free thyroxine (FT4), triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c), and high-density lipoprotein cholesterol (HDL-c) were determined. At the end of this protocol, rats were sacrificed and the thyroid glands were isolated, weighed, and examined histopathologically. The protein expression of Bcl-2 was measured by immunochemical staining. The mRNA expression of thyroglobulin (Tg), type-1 deiodinase (D1), and thyroid peroxidase (TPO) were detected by polymerase chain reaction (PCR). RESULTS: Compared with the NC group, FT3 and FT4 levels in the DRT and DC groups significantly increased at week 4 but declined thereafter. The AMT and AC groups had lower FT3 levels but comparable FT4 levels. The levels of TG, LDL-c, and HDL-c in the NC group were lower than those in the other groups whereas the LDL-c/HDL-c ratio was lowest in the AMT group. Bcl-2 expression significantly increased in the DRT group. The mRNA expression of Tg increased whereas the mRNA expression of D1 decreased. Dronedarone induced hyperthyroidism at the early stage and hypothyroidism at the late stage whereas amiodarone only caused hypothyroidism. CONCLUSION: Both dronedarone and amiodarone can induce dyslipidemia and increase the levels of TC, LDL-c, and HDL-c, and these effects may be associated with thyroid dysfunction.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/adverse effects , Anti-Arrhythmia Agents/adverse effects , Dyslipidemias/chemically induced , Thyroid Gland/drug effects , Thyroid Gland/pathology , Vasodilator Agents/adverse effects , Animals , Dronedarone , Dyslipidemias/blood , Dyslipidemias/metabolism , Dyslipidemias/pathology , Lipid Metabolism/drug effects , Lipids/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Proto-Oncogene Proteins c-bcl-2/analysis , Rats, Sprague-Dawley , Thyroid Gland/metabolism , Thyroxine/blood , Thyroxine/metabolism , Triiodothyronine/blood , Triiodothyronine/metabolism
18.
Biopharm Drug Dispos ; 37(6): 345-51, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27194397

ABSTRACT

The objectives of the current study were to characterize the pharmacokinetic profile of dronedarone in the rat, and to examine the effect of hyperlipidemia on its pharmacokinetics. Single doses of dronedarone were administered to rats intravenously (4 mg/kg), orally (55 mg/kg) and intraperitoneally (65 mg/kg). To induce hyperlipidemia, some of the rats were administered intraperitoneal doses of poloxamer 407 before giving an oral dose of dronedarone. After intravenous doses of 4 mg/kg dronedarone, plasma clearance and volume of distribution at steady-state were 25.1 ± 8.09 mL/min/kg and 10.8 ± 4.77 L/kg, respectively. After oral doses the maximum plasma concentrations (Cmax) and their median time of attainment (tmax) were 1.87 ± 1.65 mg/mL and 3.5 h, respectively. Intraperitoneal administration of 65 mg/kg dronedarone base yielded plasma Cmax and median tmax of 0.816 ± 0.611 mg/mL and 3 h, respectively. Protein binding was high in NL and HL plasma. Dronedarone is extensively distributed with high volume of distribution in the rat. The drug showed poor bioavailability (<20%) after oral and intraperitoneal administration. The increased plasma concentrations after oral dosing to hyperlipidemic rats appears to be attributable to a direct effect on metabolizing enzymes or transport proteins. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Amiodarone/analogs & derivatives , Anti-Arrhythmia Agents/pharmacokinetics , Hyperlipidemias/metabolism , Administration, Intravenous , Administration, Oral , Amiodarone/administration & dosage , Amiodarone/blood , Amiodarone/pharmacokinetics , Animals , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/blood , Area Under Curve , Biological Availability , Dronedarone , Hyperlipidemias/chemically induced , Injections, Intraperitoneal , Male , Poloxamer , Protein Binding , Rats, Sprague-Dawley
19.
Chem Pharm Bull (Tokyo) ; 64(8): 1149-53, 2016.
Article in English | MEDLINE | ID: mdl-27477654

ABSTRACT

We have developed a convergent synthesis of dronedarone, an antiarrhythmic agent. The key steps of the process are the construction of a benzofuran skeleton by iodocyclization and the carbonylative Suzuki-Miyaura cross-coupling for biaryl ketone formation. This synthetic route required only eight steps from 2-amino-4-nitrophenol in 23% overall yield.


Subject(s)
Amiodarone/analogs & derivatives , Anti-Arrhythmia Agents/chemical synthesis , Amiodarone/chemical synthesis , Amiodarone/chemistry , Anti-Arrhythmia Agents/chemistry , Dronedarone , Molecular Structure
20.
Drug Metab Dispos ; 43(4): 510-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25609219

ABSTRACT

Clopidogrel is a prodrug that needs to be converted to its active metabolite (clopi-H4) in two sequential cytochrome P450 (P450)-dependent steps. In the present study, a dynamic physiologically based pharmacokinetic (PBPK) model was developed in Simcyp for clopidogrel and clopi-H4 using a specific sequential metabolite module in four populations with phenotypically different CYP2C19 activity (poor, intermediate, extensive, and ultrarapid metabolizers) receiving a loading dose of 300 mg followed by a maintenance dose of 75 mg. This model was validated using several approaches. First, a comparison of predicted-to-observed area under the curve (AUC)0-24 obtained from a randomized crossover study conducted in four balanced CYP2C19-phenotype metabolizer groups was performed using a visual predictive check method. Second, the interindividual and intertrial variability (on the basis of AUC0-24 comparisons) between the predicted trials and the observed trial of individuals, for each phenotypic group, were compared. Finally, a further validation, on the basis of drug-drug-interaction prediction, was performed by comparing observed values of clopidogrel and clopi-H4 with or without dronedarone (moderate CYP3A4 inhibitor) coadministration using a previously developed and validated physiologically based PBPK dronedarone model. The PBPK model was well validated for both clopidogrel and its active metabolite clopi-H4, in each CYP2C19-phenotypic group, whatever the treatment period (300-mg loading dose and 75-mg last maintenance dose). This is the first study proposing a full dynamic PBPK model able to accurately predict simultaneously the pharmacokinetics of the parent drug and of its primary and secondary metabolites in populations with genetically different activity for a metabolizing enzyme.


Subject(s)
Cytochrome P-450 CYP2C19/genetics , Models, Biological , Polymorphism, Single Nucleotide , Secondary Metabolism/physiology , Ticlopidine/analogs & derivatives , Adolescent , Adult , Aged , Amiodarone/administration & dosage , Amiodarone/analogs & derivatives , Amiodarone/pharmacokinetics , Area Under Curve , Biotransformation , Clopidogrel , Cross-Over Studies , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C19 Inhibitors/pharmacology , Double-Blind Method , Dronedarone , Drug Interactions , Humans , Intestinal Absorption , Male , Middle Aged , Reproducibility of Results , Ticlopidine/administration & dosage , Ticlopidine/metabolism , Ticlopidine/pharmacokinetics , Tissue Distribution , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL