Skip to main content
Erschienen in: Journal of Interventional Cardiac Electrophysiology 2/2015

Open Access 01.03.2015

A placebo-controlled, double-blind, randomized, multicenter study to assess the effects of dronedarone 400 mg twice daily for 12 weeks on atrial fibrillation burden in subjects with permanent pacemakers

verfasst von: Michael D. Ezekowitz, Kenneth A. Ellenbogen, John P. DiMarco, Karoly Kaszala, Alexander Boddy, Gregory Geba P., Andrew Koren

Erschienen in: Journal of Interventional Cardiac Electrophysiology | Ausgabe 2/2015

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Purpose

Dronedarone is a benzofuran derivative with a pharmacological profile similar to amiodarone but has a more rapid onset of action and a much shorter half-life (13–19 h). Our goal was to evaluate the efficacy of dronedarone in atrial fibrillation (AF) patients using dual-chamber pacemakers capable of quantifying atrial fibrillation burden.

Methods

Pacemakers were adjusted to optimize AF detection. Patients with AF burden >1 % were randomized to dronedarone 400 mg twice daily (BID) or placebo. Pacemakers were interrogated after 4 and 12 weeks of treatment. The primary endpoint was the change in AF burden from baseline over the 12-week treatment period. Patients with permanent AF, severe/recently decompensated heart failure, and current use of antiarrhythmic drugs were excluded. AF burden was assessed by a core laboratory blinded to treatment assignment.

Results

From 285 patients screened, 112 were randomized (mean age 76 years, 60 % male, 84 % hypertensive, 65 % with sick sinus syndrome, 26 % with diabetes mellitus type II, 15 % with heart failure). Baseline mean (SEM) AF burden was 8.77 % (0.16) for placebo and 10.14 % (0.17) for dronedarone. Over the 12-week study period, AF burden compared to baseline decreased by 54.4 % (0.22) (P = 0.0009) with dronedarone and trended higher by 12.8 % (0.16) (P = 0.450) with placebo. The absolute change in burden was decreased by 5.5 % in the dronedarone group and increased by 1.1 % in the placebo group. Heart rate during AF was reduced to approximately 4 beats/min with dronedarone (P = 0.285). Adverse events were higher with dronedarone compared to placebo (65 vs 56 %).

Conclusions

Dronedarone reduced pacemaker-assessed the relative AF burden compared to baseline and placebo by over 50 % during the 12-week observation period.
Hinweise
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1007/​s10840-015-0001-9.

1 Introduction

There is a need to develop safe and effective antiarrhythmic agents for the treatment of atrial fibrillation (AF) [1]. Dronedarone HCl, a benzofuran derivative with a pharmacologic profile similar to amiodarone [25], was approved by the Food and Drug Administration in 2009. In the USA, it is indicated to reduce the risk of hospitalization for AF in patients in sinus rhythm with a history of paroxysmal or persistent AF [6]. Results from key trials involving nearly 6000 patients demonstrated that dronedarone reduces AF recurrence and the composite of CV hospitalization and death in non-permanent AF [7, 8]. Dronedarone possesses all four Vaughan-Williams antiarrhythmic properties, with blockade of sodium channels, noncompetitive antiadrenergic activity, prolongation of the action potential and refractory periods, and calcium antagonist properties [2, 5, 6]. Compared to amiodarone, dronedarone lacks iodine, is less lipophilic, and has a shorter half-life (13–19 h vs ~58 days for amiodarone), thereby decreasing accumulation in tissue [5, 6, 9]. Dronedarone has a low systemic bioavailability, which increases up to 15 % when the drug is administered after a fatty meal. After repeated administration of 400 mg twice daily (BID), steady state is reached within 4 to 8 days [6, 10]. Dronedarone is metabolized by CYP3A enzyme system [6, 10].
The efficacy of antiarrhythmic drugs in patients with AF has been tested using the time to first recurrence of symptomatic AF [1114]; AF events, however, frequently occur in clusters, thus questioning the validity of this endpoint [1517]. In addition, time to first recurrence data fail to quantify AF burden (i.e., percent of time in AF), frequency, and number of episodes [12], all of which may carry a different prognosis. Asymptomatic episodes are common and complicate the assessment and may in fact represent the majority of AF episodes and may be associated with mortality and stroke risk [18, 19]. The availability of continuous monitoring permits documentation of frequency, duration, time of onset, and rate of AF as well as AF burden [17, 20, 21]. Patients with accepted indications for permanent pacemakers, particularly patients with sinus node disease, are at significant risk for AF, with prevalence rates of up to and over 50 % in some studies [2224]. The Effects of Dronedarone on Atrial Fibrillation Burden in Subjects with Permanent Pacemakers (HESTIA; ClinicalTrials.gov number, NCT01135017) evaluates the impact of dronedarone on AF burden in patients with AF and permanent cardiac pacemakers.

2 Methods

The intent of HESTIA was to enroll 290 AF patients with an interim analysis when 150 patients completed. Recruitment was slow and the study was terminated after enrollment of 112 patients. For entry, patients had to be at least 21 years of age with documented AF and sinus rhythm within the previous 6 months. Patients were included if one of the following CV risk factors were present: age ≥70 years, hypertension, diabetes mellitus, prior CV accident (stroke or transient ischemic attack) or systemic embolism, or M-mode or 2D echocardiography findings showing a left atrium diameter ≥50 mm or left ventricular ejection fraction less than ≤0.40 within the prior 12 months.
HESTIA was a placebo-controlled, double-blind, randomized, multicenter trial designed to assess the effects of dronedarone 400 mg BID versus placebo on AF burden in patients with a permanent pacemaker. Following patient consent, pacemakers were programmed to optimize atrial sensing. First, far-field R wave oversensing at maximum atrial sensitivity was assessed both during ventricular pacing and during ventricular sensing (if feasible). If far-field R wave oversensing was present, post-ventricular atrial blanking period (PVAB) was increased 25 ms beyond the longest far-field R wave signal. If there was no far-field R wave oversensing, PVAB was programmed to nominal setting. Final atrial sensitivity was set between 0.25 to 0.35 mV or above if needed (minimum allowable sensitivity was 0.75 mV). Patients were excluded if atrial sensing was <1 mV. The pacemakers were programmed to DDD or MVP mode and ventricular sensing was promoted by programming long AV delay if intrinsic conduction allowed. Mode switch rate was set to 200 bpm and the maximum number of bipolar atrial electrograms (EGMs) was stored for these episodes in the pacemakers. EGMs were reviewed as a quality control measure to ensure that they were due to atrial arrhythmias and that mode switches were accurate. Atrial overdrive pacing or atrial tachycardia therapies were programmed off. The study was conducted with support of a Pacemaker Core Laboratory (PERFUSE, Boston, MA) to ensure quality control. The core laboratory reviewed all interrogation reports and electrograms to ensure that pacemaker settings were consistent with protocol requirements and that pacemakers were accurately recording AF (i.e., atrial high rate episodes).
Patients were allowed treatment with concomitant medications based on dronedarone prescribing recommendations. Where possible, digoxin was either discontinued or halved. Treatment with Vaughan-Williams class I and III antiarrhythmics was prohibited. Use of amiodarone was prohibited within 4 weeks of screening.
Patients were randomized 1:1 to dronedarone or placebo and treated for 12 weeks. The trial was conducted in accordance with all applicable ethical and good clinical practice principles, including the Declaration of Helsinki. Written informed consent was obtained from all participants.
All subjects had a dual-chamber pacemaker placed for an approved clinical indication from which AF burden was determined. There was an initial 4-week screening period (Fig. 1). AF burden was defined as arrhythmia time/24 h (e.g., 6 h of AF arrhythmia time would be 25 %). The AF burden had to be at least 1 % during the 4-week screening period off anti-arrhythmic therapy for study eligibility. For eligible patients, the AF burden during the screening period constituted the baseline AF burden. Pacemaker interrogations were also performed at 4 and 12 weeks after randomization. A final follow-up visit was performed 14 weeks after randomization.
The primary objective of this study was the effect of dronedarone on AF burden. Secondary objectives included the effects of dronedarone on ventricular rate during AF; patient-perceived AF burden; and symptom severity as reported by patients using the Atrial Fibrillation Severity Scale (AFSS). The number of electrical cardioversions and manual overdrive pacing events during treatment were collected.
An independent Data Monitoring Committee monitored safety, deaths, hospitalizations, and adverse events leading to drug discontinuation. Areas of special interest were congestive heart failure (CHF), diagnosis of interstitial lung disease, skin disorders, and peripheral neuropathy, the latter three are well known complications of amiodarone. CHF was assessed at screening and throughout the study. The planned sample size of 290 was estimated to have 70 % power to detect a reduction in mean AF burden of 30 % relative to the placebo group. The early termination of the study at 112 patients randomized reduced the power to detect treatment effects to under 50 %. The modified intent-to-treat (mITT) population included all patients who were randomized, treated with ≥1 dose of dronedarone, and had ≥1 post-baseline assessment of AF burden. The safety population was defined as the randomized population who received ≥1 dose of study medication. AF burden during the 12-week treatment period was analyzed in log-scale with adjustment for the baseline to compare AF burden between the dronedarone and placebo arms due to the non-normal distribution of AF burden. An analysis of covariance (ANCOVA) model was applied to the log-transformed AF burden data. The difference between treatment arms was estimated by the least squares mean method and was significant if the two-sided p value of the test was <0.05.
For secondary efficacy analyses, AF burden in the first 4 weeks and after 4 weeks of treatment was analyzed using the statistical method described for the primary efficacy variable and the average ventricular rate during AF episodes and the Atrial Fibrillation Severity Scale (AFSS) scores were analyzed without transformation using an ANCOVA model with treatment arm as a fixed effect term and the baseline value as a covariate. Electrical cardioversion (or overdrive pacing) was recorded.

3 Results

Two hundred and eighty-five subjects were screened; 112 (Fig. 2 and Table 1) were randomized to receive treatment. Cardiovascular history of randomized subjects included hypertension in 84 %, sick sinus syndrome in 65 %, and congestive heart failure in 15 % of patients (Table 2). At randomization, 82 % were prescribed rate-lowering medications and 73 % oral anticoagulants (Table 3). Two subjects did not have a post-randomization interrogation of their pacemaker. Both were excluded from the primary efficacy analysis.
Table 1
Patient baseline demographics
 
Placebo (n = 55)
Dronedarone (n = 57)
All (N = 112)
Age, mean (SD), years
74.5 (9.5)
77.3 (8.6)
75.9 (9.1)
Men, n (%)
38 (69.1)
29 (50.9)
67 (59.8)
Hispanic, n (%)
0
1 (1.8)
1 (0.9)
Race, n (%)
 Caucasian/White
50 (90.9)
54 (94.7)
104 (92.9)
 Black
5 (9.1)
2 (3.5)
7 (6.3)
 Asian/Oriental
0
1 (1.8)
1 (0.9)
Body mass index ≥30 kg/m2
24 (43.6)
22 (38.6)
46 (41.1)
Creatinine clearance, mean (SD), mL/min
73.2 (27.2)
66.1 (31.4)
69.6 (29.5)
SD standard deviation
Table 2
Cardiovascular history of randomized patients
 
Placebo (n = 55)
Dronedarone (n = 57)
All (N = 112)
Hypertension
45 (81.8)
49 (86.0)
94 (83.9)
Sick sinus syndrome
37 (67.3)
36 (63.2)
73 (65.2)
Coronary artery disease
24 (43.6)
16 (28.1)
40 (35.7)
Syncope
15 (27.3)
20 (35.1)
35 (31.3)
Atrioventricular block above first degree
13 (23.6)
9 (15.8)
22 (19.6)
Congestive heart failure
6 (10.9)
11 (19.3)
17 (15.2)
Non-rheumatic valvular heart disease
7 (12.7)
9 (15.8)
16 (14.3)
Myocardial infarction
9 (16.4)
6 (10.5)
15 (13.4)
Ischemic stroke
6 (10.9)
5 (8.8)
11 (9.8)
Peripheral arterial disease
7 (12.7)
3 (5.3)
10 (8.9)
Transient ischemic attack
5 (9.1)
3 (5.3)
8 (7.1)
Supra-ventricular tachycardia other than atrial fibrillation/atrial flutter
3 (5.5)
4 (7.0)
7 (6.3)
Deep vein thrombosis
3 (5.5)
3 (5.3)
6 (5.4)
Stroke of unknown origin
1 (1.8)
4 (7.0)
5 (4.5)
Ischemic dilated cardiomyopathy
2 (3.6)
2 (3.5)
4 (3.6)
Torsades de pointe
1 (1.8)
0
1 (0.9)
Ventricular fibrillation
1 (1.8)
0
1 (0.9)
All data are expressed as n (%)
Table 3
Summary of baseline medications
 
Placebo (n = 55)
Dronedarone (n = 57)
All (N = 112)
Beta blocking agents (except sotalol)
35 (63.6)
40 (70.2)
75 (67.0)
Angiotensin-converting enzyme inhibitors or angiotensin II receptor antagonists
33 (60.0)
24 (42.1)
57 (50.9)
Oral anticoagulant
38 (69.1)
44 (77.2)
82 (73.2)
Diuretics
25 (45.5)
21 (36.8)
46 (41.1)
Aspirin
26 (47.3)
19 (33.3)
45 (40.2)
Statins
34 (61.8)
32 (56.1)
66 (58.9)
Calcium antagonists with heart rate lowering effects
12 (21.8)
8 (14.0)
20 (17.9)
Digitalis
12 (21.8)
10 (17.5)
22 (19.6)
Other chronic antiplatelet therapy
10 (18.2)
5 (8.8)
15 (13.4)
All data are expressed as n (%)
Median duration of treatment was 12 weeks. Ninety-four subjects completed the study, and 18 subjects discontinued treatment due to the investigator’s or subject’s decision. Discontinuations due to an adverse event in the dronedarone group (14 %) exceeded those in the placebo group (6 %).
Over the 12-week treatment period, mean AF burden increased from 8.8 % (0.16) to 9.9 % (0.20) (increase of 12.8 % (0.16); P = 0.450) in the placebo group and decreased from 10.1 % (0.17) to 4.6 % (0.29) (decrease of 54.4 % (0.22); P = 0.0009) in the dronedarone group. The absolute changes in AF burden from baseline increased by 1.1 % in the placebo group and decreased by 5.5 % in the dronedarone group. Compared to placebo, AF burden in the dronedarone group was decreased by 59.1 % (0.28 %) (P = 0.0015; Fig. 3). AF burden changes with dronedarone compared to placebo in secondary efficacy analyses were consistent with the overall result (weeks 1–4, 63.2 % (0.29) reduction, P = 0.0009; weeks 5–12, 60.3 % (0.30) reduction, P = 0.003). The distribution of baseline versus weeks 1–12 AF burden in treated patients is shown in Fig. 4.
The mean ventricular rate during AF was approximately 4 bpm lower in the dronedarone treatment group compared to placebo (P = 0.285). AFSS changes from baseline were not significantly different between groups (Table 4). Only one patient in the placebo group was cardioverted.
Table 4
Summary of Atrial Fibrillation Severity Scale (AFSS)
 
Placebo (N = 55)
Dronedarone (N = 55)
AF burden
 Baseline mean (SD)
13.72 (4.59)
12.76 (4.62)
 Post-baseline mean (SD)
13.30 (5.40)
12.48 (5.45)
 Change from baseline, mean (SD)
−1.04 (3.22)
−0.68 (4.71)
 Treatment difference, LS mean (SE)a
0.14 (0.981)
   P value
0.8902
AF symptoms
 Baseline mean (SD)
8.47 (6.97)
8.36 (6.83)
 Post-baseline mean (SD)
8.06 (7.11)
7.42 (7.48)
 Change from baseline, mean (SD)
−0.28 (4.88)
−0.72 (5.66)
 Treatment difference, LS mean (SE)a
−0.49 (0.980)
   P value
0.6216
aFrom ANCOVA model
Treatment-emergent adverse events were common in both groups (dronedarone, 64.9 %; placebo, 56.4 %); most were gastrointestinal (Table 5). Treatment-emergent serious adverse events occurred in 14 patients (7 in each treatment group). Serious adverse events in the dronedarone group included congestive heart failure, peripheral neuropathy, ocular discomfort, presyncope, muscle rupture, and cardiomyopathy. More patients in the dronedarone group discontinued treatment due to adverse events compared with placebo (8 [14.0 %] vs 3 [5.5 %], respectively). There was no evidence of dronedarone-induced ventricular tachycardia and there were no deaths, strokes, or major bleeding events in the study.
Table 5
Treatment-emergent adverse events
 
Placebo (n = 55)
Dronedarone (n = 57)
Overview of treatment-emergent adverse events
 Patients with any treatment-emergent adverse event
31 (56.4)
37 (64.9)
 Patients with any treatment-emergent serious adverse event
7 (12.7)
7 (12.3)
 Patients with any treatment-emergent adverse event leading to death
0
0
 Patients with any treatment-emergent adverse event leading to permanent treatment discontinuation
3 (5.5)
8 (14.0)
Treatment-emergent adverse event of special interest
 Congestive heart failure
2 (3.6)
3 (5.3)
Treatment-emergent adverse events by primary system organ class
 Gastrointestinal disorders
10 (18.2)
17 (29.8)
 Respiratory, thoracic, and mediastinal disorders
7 (12.7)
8 (14.0)
 Nervous system disorders
6 (10.9)
7 (12.3)
 Infections and infestations
8 (14.5)
6 (10.5)
 Cardiac disorders
3 (5.5)
5 (8.8)
 Metabolism and nutrition disorders
3 (5.5)
3 (5.3)
 Vascular disorders
2 (3.6)
3 (5.3)
 Psychiatric disorders
2 (3.6)
1 (1.8)
 Eye disorders
1 (1.8)
1 (1.8)
 Ear and labyrinth disorders
2 (3.6)
1 (1.8)
 Skin and subcutaneous tissue disorders
2 (3.6)
1 (1.8)
 Musculoskeletal and connective tissue disorders
4 (7.3)
1 (1.8)
 Renal and urinary disorders
1 (1.8)
1 (1.8)
 Immune system disorders
1 (1.8)
0
 Hepatobiliary disorders
1 (1.8)
0
All data are expressed as n (%). Treatment-emergent adverse events include all patients randomized and that received at least one dose of study drug

4 Discussion

The major finding was a 59 % relative reduction of AF burden with dronedarone compared to placebo (P = 0.0015). In the placebo group, the mean (geometric) AF burden increased from 8.8 to 9.9 % during the treatment period, whereas in the dronedarone group, the mean (geometric) AF burden was decreased from 10.1 to 4.6 %, an absolute reduction of 5.5 %. The effect was consistent over the 12-week duration of treatment. Average ventricular rate during AF was slightly (4 bpm) lower in the dronedarone group, but these differences were not statistically significant. There were no statistically significant differences in the AFSS. One patient was cardioverted.
Consistent with the known profile of dronedarone, there was an increase in gastrointestinal events. Serious adverse events and discontinuations due to adverse events were infrequent but occurred more often in the dronedarone group. There were no deaths in the study.
Two large multicenter, randomized, placebo-controlled studies evaluated the efficacy of dronedarone on AF recurrence [7, 8]. In the pooled analysis of the European Trial in Atrial Fibrillation or Flutter Patients Receiving Dronedarone for the Maintenance of Sinus Rhythm (EURIDIS, NCT00259428) and the American-Australian-African Trial with Dronedarone in Atrial Fibrillation or Flutter Patients for the Maintenance of Sinus Rhythm (ADONIS, NCT00259376), dronedarone significantly delayed the time to first recurrence of AF during the 12-month study period by approximately 25 % (P < 0.001 vs placebo), with an absolute difference in recurrence rate of ~11 % [8].
Use of pacemaker determined AF burden has been the subject of several prior studies and has potentially important clinical implications. A recent trial of patients with bradycardia and a history of AF receiving pacemakers indicated that total AF burden had a significant but weak correlation with cardiac hospitalization rates [25]. Although total AF burden is typically used as an endpoint in device trials such as the Atrial Therapy Efficacy and Safety Trial (ATTEST) [21, 2629], it may also be a useful endpoint for antiarrhythmic medications. The Paroxysmal Atrial Fibrillation Study with Continuous Atrial Fibrillation Logging (PASCAL, NCT00389792) trial found up to a 74 % reduction in AF burden with budiodarone in an investigational dose ranging study [17].
The study had several limitations. First, study enrollment was below goals (290 planned vs 112 actual patients with interim analysis planned at 150 patients) because of the difficulty in finding non-permanent AF patients with at least 1 % AF burden on initial screening and during the 4-week baseline period. The study, however, was analyzed as originally planned with the primary endpoint achieving a highly significant (P = 0.0015) outcome. A second limitation is the use of mode switches to assess AF burden with the risks of under or oversensing. Efforts were made to mitigate this risk including use of a pacemaker core lab to ensure that pacemakers were appropriately set to study requirements and that pacemakers were accurately recording AF. Last, because HESTIA was placebo-controlled, patients with more severe AF symptoms were excluded. Low baseline AFSS burden and symptom scores reflect this and may have mitigated finding significant changes in the AFSS despite significant reductions in AF burden observed in the study.
The HESTIA study has important implications on design of future trials of antiarrhythmic drugs for AF. A post hoc calculation of study power based on our study findings was >95 %, underscoring the sensitivity of continuous ECG monitoring for assessing AF. In comparison, the EURIDIS and ADONIS studies required >500 patients each to detect a 25 % reduction in the rate of AF recurrence (at 12 months) with dronedarone compared to placebo with 90 % power. Continuous ECG monitoring also provides a more complete characterization of AF patterns compared with traditional time to first recurrence assessments, and also offers a sensitive method of detecting ventricular arrhythmias.

5 Conclusion

The HESTIA study was designed to investigate the effects of dronedarone on AF burden in a population of patients experiencing intermittent episodes of AF. Prior to randomization, these patients were in AF an average of 10 % of the time. Dronedarone reduced the primary endpoint, time in AF, by slightly more than half (P < 0.005). This effect was consistent over the 12-week duration of treatment. Safety findings were consistent with the known profile of dronedarone.

Conflict of interest

Michael D. Ezekowitz has served as a consultant to and speaker for Boehringer Ingelheim, Ridgefield, CT, and as a consultant to Pfizer, New York, NY; Sanofi-aventis US, Bridgewater, NJ; Bristol-Myers Squibb, New York, NY; Portola Pharmaceuticals, Plumas County, CA; Bayer, Pittsburgh, PA; Daiichi Sankyo, Parsippany, NJ; Medtronic, Minneapolis, MN; Aegerion Pharmaceuticals, Cambridge, MA; Merck, Whitehouse Station, NJ; Johnson & Johnson, New Brunswick, NJ; Gilead Sciences, Inc., Foster City, CA; Janssen Scientific Affairs, Titusville, NJ; Pozen Inc, Chapel Hill, NC; and Coherex Medical, Salt Lake City, UT. Kenneth A. Ellenbogen has served as a consultant to Pfizer. John P. DiMarco has received grant/research support from Gilead Sciences, Inc., Sanofi-aventis US, has served as a consultant to Novartis Pharmaceuticals Corp., East Hanover, NJ, and St. Jude Medical, Secaucus, NJ, has received honoraria from Boston Scientific, Natick, MA, and has served as an advisor to Medtronic. Karoly Kaszala has served as a consultant to Sanofi-aventis US. Alexander Boddy and Andrew Koren are employees of Sanofi-aventis US. Gregory Geba was an employee of Sanofi-aventis US at the time this study was conducted.

Funding sources

This study was funded by Sanofi-aventis US, Bridgewater, NJ. Editorial support was provided by Peloton Advantage, LLC, Parsippany, NJ, and Paulette Trent, funded by Sanofi-aventis US. The opinions expressed in the current article are those of the authors. The authors received no honorarium or other form of financial support related to the development of this manuscript.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Kernan, W. N., Ovbiagele, B., Black, H. R., Bravata, D. M., Chimowitz, M. I., Ezekowitz, M. D., Fang, M. C., Fisher, M., Furie, K. L., Heck, D. V., Johnston, S. C., Kasner, S. E., Kittner, S. J., Mitchell, P. H., Rich, M. W., Richardson, D., Schwamm, L. H., Wilson, J. A., & on behalf of the American Heart Association Stroke Council, Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, and Council on Peripheral Vascular Disease. (2014). Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke, 45, 2160–2236.CrossRefPubMed Kernan, W. N., Ovbiagele, B., Black, H. R., Bravata, D. M., Chimowitz, M. I., Ezekowitz, M. D., Fang, M. C., Fisher, M., Furie, K. L., Heck, D. V., Johnston, S. C., Kasner, S. E., Kittner, S. J., Mitchell, P. H., Rich, M. W., Richardson, D., Schwamm, L. H., Wilson, J. A., & on behalf of the American Heart Association Stroke Council, Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, and Council on Peripheral Vascular Disease. (2014). Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke, 45, 2160–2236.CrossRefPubMed
2.
Zurück zum Zitat Gautier, P., Guillemare, E., Marion, A., Bertrand, J. P., Tourneur, Y., & Nisato, D. (2003). Electrophysiologic characterization of dronedarone in guinea pig ventricular cells. Journal of Cardiovascular Pharmacology, 41, 191–202.CrossRefPubMed Gautier, P., Guillemare, E., Marion, A., Bertrand, J. P., Tourneur, Y., & Nisato, D. (2003). Electrophysiologic characterization of dronedarone in guinea pig ventricular cells. Journal of Cardiovascular Pharmacology, 41, 191–202.CrossRefPubMed
3.
Zurück zum Zitat Hodeige, D., Heyndrickx, J. P., Chatelain, P., & Manning, A. (1995). SR 33589, a new amiodarone-like antiarrhythmic agent: anti-adrenoceptor activity in anaesthetized and conscious dogs. European Journal of Pharmacology, 279, 25–32.CrossRefPubMed Hodeige, D., Heyndrickx, J. P., Chatelain, P., & Manning, A. (1995). SR 33589, a new amiodarone-like antiarrhythmic agent: anti-adrenoceptor activity in anaesthetized and conscious dogs. European Journal of Pharmacology, 279, 25–32.CrossRefPubMed
4.
Zurück zum Zitat Sun, W., Sarma, J. S., & Singh, B. N. (1999). Electrophysiological effects of dronedarone (SR33589), a noniodinated benzofuran derivative, in the rabbit heart: comparison with amiodarone. Circulation, 100, 2276–2281.CrossRefPubMed Sun, W., Sarma, J. S., & Singh, B. N. (1999). Electrophysiological effects of dronedarone (SR33589), a noniodinated benzofuran derivative, in the rabbit heart: comparison with amiodarone. Circulation, 100, 2276–2281.CrossRefPubMed
5.
Zurück zum Zitat Wegener, F. T., Ehrlich, J. R., & Hohnloser, S. H. (2006). Dronedarone: an emerging agent with rhythm- and rate-controlling effects. Journal of Cardiovascular Electrophysiology, 17(Suppl 2), S17–S20.CrossRefPubMed Wegener, F. T., Ehrlich, J. R., & Hohnloser, S. H. (2006). Dronedarone: an emerging agent with rhythm- and rate-controlling effects. Journal of Cardiovascular Electrophysiology, 17(Suppl 2), S17–S20.CrossRefPubMed
6.
Zurück zum Zitat (2012). Multaq [package insert]. Bridgewater, NJ: sanofi-aventis U.S. LLC. (2012). Multaq [package insert]. Bridgewater, NJ: sanofi-aventis U.S. LLC.
7.
Zurück zum Zitat Hohnloser, S. H., Crijns, H. J., van Eickels, M., Gaudin, C., Page, R. L., Torp-Pedersen, C., & Connolly, S. J. (2009). Effect of dronedarone on cardiovascular events in atrial fibrillation. New England Journal of Medicine, 360, 668–678.CrossRefPubMed Hohnloser, S. H., Crijns, H. J., van Eickels, M., Gaudin, C., Page, R. L., Torp-Pedersen, C., & Connolly, S. J. (2009). Effect of dronedarone on cardiovascular events in atrial fibrillation. New England Journal of Medicine, 360, 668–678.CrossRefPubMed
8.
Zurück zum Zitat Singh, B. N., Connolly, S. J., Crijns, H. J., Roy, D., Kowey, P. R., Capucci, A., Radzik, D., Aliot, E. M., & Hohnloser, S. H. (2007). Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter. New England Journal of Medicine, 357, 987–999.CrossRefPubMed Singh, B. N., Connolly, S. J., Crijns, H. J., Roy, D., Kowey, P. R., Capucci, A., Radzik, D., Aliot, E. M., & Hohnloser, S. H. (2007). Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter. New England Journal of Medicine, 357, 987–999.CrossRefPubMed
9.
Zurück zum Zitat (2008). Pacerone [package insert]. Minneapolis, MN: Upsher-Smith Laboratories, Inc. (2008). Pacerone [package insert]. Minneapolis, MN: Upsher-Smith Laboratories, Inc.
10.
Zurück zum Zitat Krishnamoorthy, S., & Lip, G. Y. (2009). Antiarrhythmic drugs for atrial fibrillation: focus on dronedarone. Expert Review of Cardiovascular Therapy, 7, 473–481.CrossRefPubMed Krishnamoorthy, S., & Lip, G. Y. (2009). Antiarrhythmic drugs for atrial fibrillation: focus on dronedarone. Expert Review of Cardiovascular Therapy, 7, 473–481.CrossRefPubMed
11.
Zurück zum Zitat Wyse, D. G. (2004). Overview of endpoints in atrial fibrillation studies. Heart Rhythm, 1, B3–7. discussion.CrossRefPubMed Wyse, D. G. (2004). Overview of endpoints in atrial fibrillation studies. Heart Rhythm, 1, B3–7. discussion.CrossRefPubMed
12.
Zurück zum Zitat Camm, A. J., & Reiffel, J. A. (2008). Defining endpoints in clinical trials on atrial fibrillation. European Heart Journal, 10(Suppl H), H55–H78.CrossRef Camm, A. J., & Reiffel, J. A. (2008). Defining endpoints in clinical trials on atrial fibrillation. European Heart Journal, 10(Suppl H), H55–H78.CrossRef
13.
Zurück zum Zitat Pritchett, E. L. (2004). Symptomatic arrhythmia recurrence as an outcome in clinical trials of antiarrhythmic drug therapy. Heart Rhythm, 1, B36–B40.CrossRefPubMed Pritchett, E. L. (2004). Symptomatic arrhythmia recurrence as an outcome in clinical trials of antiarrhythmic drug therapy. Heart Rhythm, 1, B36–B40.CrossRefPubMed
14.
Zurück zum Zitat Singh, B. N., Singh, S. N., Reda, D. J., Tang, X. C., Lopez, B., Harris, C. L., Fletcher, R. D., Sharma, S. C., Atwood, J. E., Jacobson, A. K., Lewis, H. D., Jr., Raisch, D. W., & Ezekowitz, M. D. (2005). Amiodarone versus sotalol for atrial fibrillation. New England Journal of Medicine, 352, 1861–1872.CrossRefPubMed Singh, B. N., Singh, S. N., Reda, D. J., Tang, X. C., Lopez, B., Harris, C. L., Fletcher, R. D., Sharma, S. C., Atwood, J. E., Jacobson, A. K., Lewis, H. D., Jr., Raisch, D. W., & Ezekowitz, M. D. (2005). Amiodarone versus sotalol for atrial fibrillation. New England Journal of Medicine, 352, 1861–1872.CrossRefPubMed
15.
Zurück zum Zitat Kaemmerer, W. F., Rose, M. S., & Mehra, R. (2001). Distribution of patients’ paroxysmal atrial tachyarrhythmia episodes: implications for detection of treatment efficacy. Journal of Cardiovascular Electrophysiology, 12, 121–130.CrossRefPubMed Kaemmerer, W. F., Rose, M. S., & Mehra, R. (2001). Distribution of patients’ paroxysmal atrial tachyarrhythmia episodes: implications for detection of treatment efficacy. Journal of Cardiovascular Electrophysiology, 12, 121–130.CrossRefPubMed
16.
Zurück zum Zitat Shehadeh, L. A., Liebovitch, L. S., & Wood, M. A. (2002). Temporal patterns of atrial arrhythmia recurrences in patients with implantable defibrillators: implications for assessing antiarrhythmic therapies. Journal of Cardiovascular Electrophysiology, 13, 303–309.CrossRefPubMed Shehadeh, L. A., Liebovitch, L. S., & Wood, M. A. (2002). Temporal patterns of atrial arrhythmia recurrences in patients with implantable defibrillators: implications for assessing antiarrhythmic therapies. Journal of Cardiovascular Electrophysiology, 13, 303–309.CrossRefPubMed
17.
Zurück zum Zitat Ezekowitz, M. D., Nagarakanti, R., Lubinski, A., Bandman, O., Canafax, D., Ellis, D. J., Milner, P. G., Ziola, M., Thibault, B., & Hohnloser, S. H. (2012). A randomized trial of budiodarone in paroxysmal atrial fibrillation. Journal of Interventional Cardiac Electrophysiology, 34, 1–9.CrossRefPubMed Ezekowitz, M. D., Nagarakanti, R., Lubinski, A., Bandman, O., Canafax, D., Ellis, D. J., Milner, P. G., Ziola, M., Thibault, B., & Hohnloser, S. H. (2012). A randomized trial of budiodarone in paroxysmal atrial fibrillation. Journal of Interventional Cardiac Electrophysiology, 34, 1–9.CrossRefPubMed
18.
Zurück zum Zitat Flaker, G. C., Belew, K., Beckman, K., Vidaillet, H., Kron, J., Safford, R., Mickel, M., & Barrell, P. (2005). Asymptomatic atrial fibrillation: demographic features and prognostic information from the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study. American Heart Journal, 149, 657–663.CrossRefPubMed Flaker, G. C., Belew, K., Beckman, K., Vidaillet, H., Kron, J., Safford, R., Mickel, M., & Barrell, P. (2005). Asymptomatic atrial fibrillation: demographic features and prognostic information from the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study. American Heart Journal, 149, 657–663.CrossRefPubMed
19.
Zurück zum Zitat Healey, J. S., Connolly, S. J., Gold, M. R., Israel, C. W., van Gelder, I. C., Capucci, A., Lau, C. P., Fain, E., Yang, S., Bailleul, C., Morillo, C. A., Carlson, M., Themeles, E., Kaufman, E. S., & Hohnloser, S. H. (2012). Subclinical atrial fibrillation and the risk of stroke. New England Journal of Medicine, 366, 120–129.CrossRefPubMed Healey, J. S., Connolly, S. J., Gold, M. R., Israel, C. W., van Gelder, I. C., Capucci, A., Lau, C. P., Fain, E., Yang, S., Bailleul, C., Morillo, C. A., Carlson, M., Themeles, E., Kaufman, E. S., & Hohnloser, S. H. (2012). Subclinical atrial fibrillation and the risk of stroke. New England Journal of Medicine, 366, 120–129.CrossRefPubMed
20.
Zurück zum Zitat Gillis, A. M. (2004). Rhythm control in atrial fibrillation: endpoints for device-based trials. Heart Rhythm, 1, B52–B57.CrossRefPubMed Gillis, A. M. (2004). Rhythm control in atrial fibrillation: endpoints for device-based trials. Heart Rhythm, 1, B52–B57.CrossRefPubMed
21.
Zurück zum Zitat Euler, D. E., & Friedman, P. A. (2003). Atrial arrhythmia burden as an endpoint in clinical trials: is it the best surrogate? Lessons from a multicenter defibrillator trial. Cardiac Electrophysiology Review, 7, 355–358.CrossRefPubMed Euler, D. E., & Friedman, P. A. (2003). Atrial arrhythmia burden as an endpoint in clinical trials: is it the best surrogate? Lessons from a multicenter defibrillator trial. Cardiac Electrophysiology Review, 7, 355–358.CrossRefPubMed
22.
Zurück zum Zitat Gillis, A. M., & Morck, M. (2002). Atrial fibrillation after DDDR pacemaker implantation. Journal of Cardiovascular Electrophysiology, 13, 542–547.CrossRefPubMed Gillis, A. M., & Morck, M. (2002). Atrial fibrillation after DDDR pacemaker implantation. Journal of Cardiovascular Electrophysiology, 13, 542–547.CrossRefPubMed
23.
Zurück zum Zitat Israel, C. W., Gronefeld, G., Ehrlich, J. R., Li, Y. G., & Hohnloser, S. H. (2004). Long-term risk of recurrent atrial fibrillation as documented by an implantable monitoring device: implications for optimal patient care. Journal of the American College of Cardiology, 43, 47–52.CrossRefPubMed Israel, C. W., Gronefeld, G., Ehrlich, J. R., Li, Y. G., & Hohnloser, S. H. (2004). Long-term risk of recurrent atrial fibrillation as documented by an implantable monitoring device: implications for optimal patient care. Journal of the American College of Cardiology, 43, 47–52.CrossRefPubMed
24.
Zurück zum Zitat Padeletti, L., Santini, M., Boriani, G., Botto, G., Capucci, A., Gulizia, M., Ricci, R., Spampinato, A., Pieragnoli, P., Warman, E., Vimercati, M., & Grammatico, A. (2005). Temporal variability of atrial tachyarrhythmia burden in bradycardia-tachycardia syndrome patients. European Heart Journal, 26, 165–172.CrossRefPubMed Padeletti, L., Santini, M., Boriani, G., Botto, G., Capucci, A., Gulizia, M., Ricci, R., Spampinato, A., Pieragnoli, P., Warman, E., Vimercati, M., & Grammatico, A. (2005). Temporal variability of atrial tachyarrhythmia burden in bradycardia-tachycardia syndrome patients. European Heart Journal, 26, 165–172.CrossRefPubMed
25.
Zurück zum Zitat Mehra, R., Ziegler, P., & Koehler, J. (2004). Diagnostic information in implantable devices that pertain to endpoints in atrial fibrillation studies. Heart Rhythm, 1, B64–B69.CrossRefPubMed Mehra, R., Ziegler, P., & Koehler, J. (2004). Diagnostic information in implantable devices that pertain to endpoints in atrial fibrillation studies. Heart Rhythm, 1, B64–B69.CrossRefPubMed
26.
Zurück zum Zitat Carlson, M. D., Ip, J., Messenger, J., Beau, S., Kalbfleisch, S., Gervais, P., Cameron, D. A., Duran, A., Val-Mejias, J., Mackall, J., & Gold, M. (2003). A new pacemaker algorithm for the treatment of atrial fibrillation: results of the Atrial Dynamic Overdrive Pacing Trial (ADOPT). Journal of the American College of Cardiology, 42, 627–633.CrossRefPubMed Carlson, M. D., Ip, J., Messenger, J., Beau, S., Kalbfleisch, S., Gervais, P., Cameron, D. A., Duran, A., Val-Mejias, J., Mackall, J., & Gold, M. (2003). A new pacemaker algorithm for the treatment of atrial fibrillation: results of the Atrial Dynamic Overdrive Pacing Trial (ADOPT). Journal of the American College of Cardiology, 42, 627–633.CrossRefPubMed
27.
Zurück zum Zitat Friedman, P. A., Dijkman, B., Warman, E. N., Xia, H. A., Mehra, R., Stanton, M. S., & Hammill, S. C. (2001). Atrial therapies reduce atrial arrhythmia burden in defibrillator patients. Circulation, 104, 1023–1028.CrossRefPubMed Friedman, P. A., Dijkman, B., Warman, E. N., Xia, H. A., Mehra, R., Stanton, M. S., & Hammill, S. C. (2001). Atrial therapies reduce atrial arrhythmia burden in defibrillator patients. Circulation, 104, 1023–1028.CrossRefPubMed
28.
Zurück zum Zitat Gillis, A. M., Connolly, S. J., Lacombe, P., Philippon, F., Dubuc, M., Kerr, C. R., Yee, R., Rose, M. S., Newman, D., Kavanagh, K. M., Gardner, M. J., Kus, T., & Wyse, D. G. (2000). Randomized crossover comparison of DDDR versus VDD pacing after atrioventricular junction ablation for prevention of atrial fibrillation. The atrial pacing peri-ablation for paroxysmal atrial fibrillation (PA (3)) study investigators. Circulation, 102, 736–741.CrossRefPubMed Gillis, A. M., Connolly, S. J., Lacombe, P., Philippon, F., Dubuc, M., Kerr, C. R., Yee, R., Rose, M. S., Newman, D., Kavanagh, K. M., Gardner, M. J., Kus, T., & Wyse, D. G. (2000). Randomized crossover comparison of DDDR versus VDD pacing after atrioventricular junction ablation for prevention of atrial fibrillation. The atrial pacing peri-ablation for paroxysmal atrial fibrillation (PA (3)) study investigators. Circulation, 102, 736–741.CrossRefPubMed
29.
Zurück zum Zitat Lee, M. A., Weachter, R., Pollak, S., Kremers, M. S., Naik, A. M., Silverman, R., Tuzi, J., Wang, W., Johnson, L. J., & Euler, D. E. (2003). The effect of atrial pacing therapies on atrial tachyarrhythmia burden and frequency: results of a randomized trial in patients with bradycardia and atrial tachyarrhythmias. Journal of the American College of Cardiology, 41, 1926–1932.CrossRefPubMed Lee, M. A., Weachter, R., Pollak, S., Kremers, M. S., Naik, A. M., Silverman, R., Tuzi, J., Wang, W., Johnson, L. J., & Euler, D. E. (2003). The effect of atrial pacing therapies on atrial tachyarrhythmia burden and frequency: results of a randomized trial in patients with bradycardia and atrial tachyarrhythmias. Journal of the American College of Cardiology, 41, 1926–1932.CrossRefPubMed
Metadaten
Titel
A placebo-controlled, double-blind, randomized, multicenter study to assess the effects of dronedarone 400 mg twice daily for 12 weeks on atrial fibrillation burden in subjects with permanent pacemakers
verfasst von
Michael D. Ezekowitz
Kenneth A. Ellenbogen
John P. DiMarco
Karoly Kaszala
Alexander Boddy
Gregory Geba P.
Andrew Koren
Publikationsdatum
01.03.2015
Verlag
Springer US
Erschienen in
Journal of Interventional Cardiac Electrophysiology / Ausgabe 2/2015
Print ISSN: 1383-875X
Elektronische ISSN: 1572-8595
DOI
https://doi.org/10.1007/s10840-014-9966-z

Weitere Artikel der Ausgabe 2/2015

Journal of Interventional Cardiac Electrophysiology 2/2015 Zur Ausgabe

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Dihydropyridin-Kalziumantagonisten können auf die Nieren gehen

30.04.2024 Hypertonie Nachrichten

Im Vergleich zu anderen Blutdrucksenkern sind Kalziumantagonisten vom Diyhdropyridin-Typ mit einem erhöhten Risiko für eine Mikroalbuminurie und in Abwesenheit eines RAS-Blockers auch für ein terminales Nierenversagen verbunden.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.