Article

Considerations for the Development of New Antiarrhythmic Drugs

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

Several studies over the past few years have investigated the benefits of antiarrhythmic drugs (AADs) for the treatment of atrial fibrillation (AF). Data from key studies comparing rate versus rhythm control show no evidence of superiority in terms of survival benefits with either therapeutic strategy. In terms of AADs, many of the currently available drugs are associated with safety issues, thus the relative risk-benefit ratios of each drug is a major consideration when selecting an AAD in therapy. The safety issues and complications associated with current therapeutic drug options clearly indicate the need for novel drug development in the field of AF. New approaches that have been evaluated include AADs with greater atrial selectivity and novel non-selective multichannelblocking AADs. Trials evaluating new AADs will likely need to demonstrate superior safety profiles over existing drugs. However, in light of the inconclusive results in terms of survival benefits of the major rate versus rhythm studies and the increasing use of nonpharmacological techniques, including direct-current cardioversion and ablative techniques, cardiologists are faced with questions concerning the specific direction of new AAD development to be undertaken in future.

Disclosure:John Camm has received consulting fees from Sanofi Aventis, Cardiome, ARYx, BMS, Pfizer, Boehringer Ingelheim, Astellas and Merck, research grants from Daiichi, sanofi-aventis, Servier, BMS and Pfizer and lecture fees from sanofi-aventis, St Jude Medical, Medtronic, Boston Scientific, Boehringer Ingelheim and BMS. The other authors have no conflicts of interest to declare.

Received:

Accepted:

Correspondence Details:John Camm, BHF Professor of Clinical Cardiology, Cardiac and Vascular Sciences, St George's university of London, Cranmer Terrace, London, SW17 0RE, uK. E: jcamm@sgul.ac.uk

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Atrial fibrillation (AF) is the most common cardiac arrhythmia, having an estimated combined prevalence of 6.8 million in the EU and the US.1,2 It is characterised by unco-ordinated atrial activation followed by the deterioration of mechanical function, which results in disturbances to sinus rhythm and ventricular rate.2 An increased risk of heart failure (HF), thrombo-embolism, cardiomyopathy, a five-fold increased risk of stroke and an approximately two-fold increased risk of mortality have been associated with AF.2,3 AF also represents a major economic burden as it can lead to hospitalisation,1 with 70% of the cost of AF management driven by inpatient care and interventional procedures.4

Antiarrhythmic drugs (AADs) are categorised into four broad categories (I, II, III and IV), based on their dominant electrophysiological properties.5 Pharmacological cardioversion is predominantly performed with Vaughan-Williams class Ia, Ic and III AADs. Class Ic, beta-blockers or class III AADs are commonly used to maintain sinus rhythm. However, the available class Ia, Ic and III AADs have limitations to varying degrees, such as moderate efficacy and proarrhythmic or extra-cardiac toxic effects in patients with structural heart disease.6,7

This article reviews current approaches to the development of new AADs and attempts to address some of the concerns in terms of their development for the management of AF.

Atrial-selective versus non-selective Multichannel-blocking Antiarrhythmic Drugs

There are a number of different approaches in the development of new AADs for the treatment of AF. These approaches include novel non-selective multichannel-blocking drugs and atrial-specific drugs.

Rationale for the Use of Atrial-selective Drugs

It is known that different ionic currents are expressed in the atria than in the ventricles (see Figure 1).8 Atrial-selective drugs only act on these atrial-specific ionic channels, and thus theoretically avoid drug effects (including pro-arrhythmic effects) on the ventricular myocardium. Vernakalant is a novel amino-cyclohexyl ether drug that has relatively atrial-selective blocking activity. It is currently undergoing regulatory review for the indication of converting AF to sinus rhythm. Vernakalant has a unique ion-channel-blocking profile, being capable of blocking IKACh channels, early activating K+ channels (IKur, Ito) and frequencyand voltage-dependent INa channels.9 Such relatively specific blocking activity leads to an atrial-selective action potential duration (APD) and effective refractory period (ERP) prolongation. In addition, the intravenous formulation of vernakalant has exhibited a good safety profile in clinical trials, and no proarrhythmic effects were observed during or up to 90 minutes after drug infusion.10 In the Atrial Arrhythmia Conversion Trials (ACT) I,11 ACT II12 and ACT III13 and the single-arm open-label ACT IV14 trials the conversion rate from acute AF to normal sinus rhythm was significantly higher with an intravenous infusion of vernakalant versus placebo (47.0-51.7% versus 3.6-14%, respectively; p<0.001). The effects of vernakalant were durable up to 24 hours. These data are consistent with the results of the Controlled Randomised Atrial Fibrillation Trial (CRAFT) phase II study in which the conversion rate from acute AF to normal sinus rhythm was 52.9% with vernakalant versus 5.3% for placebo (p=0.0014).15 Importantly, the median time to sinus rhythm conversion was shown to be 11 minutes in the ACT I trial, and such a rapid conversion effect has implications for the clinical use of this drug.11 Vernakalant infusion was associated with QT prolongations, but no cases of torsade de pointes were reported. Vernakalant was also associated with a higher occurrence of moderate to severe hypotension (2.6 versus and 0.6% on placebo). The atrial-selective AVE 0118 blocks the early activating K+ channels (IKur, Ito). Preliminary studies with the agent showed effects on atrial contraction in animal models without proarrhythmic effects on the ventricle.16 Studies on isolated atrial myocardium from patients with AF showed that AVE 0118 enhanced atrial contractility. The positive inotropic effect was atrial-specific and was shown to be due to the changes of the action potential configuration that enhanced Ca2+ entry via reverse mode Na+/Ca2+ exchange.17 The development of the drug has since been stopped.

Rationale for the Use of Non-selective Multichannel-blocking Drugs

Non-selective multichannel-blocking drugs act to inhibit not only the ionic channels in the atria but also other specific and non-specific channels. Such activity may provide additional benefits compared with atrial-specific drugs, i.e. beta-receptor blockade. The most widely used non-selective multichannel-blocking drug is the class III amiodarone. It is a highly effective treatment for AF but is associated with several adverse events, especially extra-cardiac toxicities.6 More recently, a novel orally administered non-selective multichannelblocking drug, dronedarone, has been developed. Dronedarone has recently been approved by the US Food and Drug Administration (FDA) for reducing the risk of cardiovascular hospitalisation in patients with paroxysmal/ persistent AF or atrial flutter (AFL). This group of patients are required to have had a recent episode of AF/AFL, to have associated cardiovascular risk factors and to be in sinus rhythm or to be scheduled for cardioversion. Dronedarone is contraindicated in patients with New York Heart Association (NYHA) class IV HF or NYHA class II-III HF with a recent decompensation requiring hospitalisation or referral to a specialised HF clinic. In the EU, dronedarone is indicated in clinically stable adult patients with a history of or current non-permanent AF to prevent recurrence of AF or to lower the ventricular rate. Like amiodarone, dronedarone can block Na+, Ca+ and multiple K+ channels to produce a sympatholytic blockade.18 Dronedarone is a structurally modified version of amiodarone, and these structural changes offer the former some advantages. The presence of a methane-sulfonyl group to amiodarone results in lowering of dronedarone's lipophilicity and, consequently, a reduction in its half-life.19 This helps reduce the toxic effects of long-term dronedarone administration. The addition of the methane-sulfonyl group and the removal of iodine from amiodarone leads to a reduction in the risk of thyroid-related effects and pulmonary fibrosis.19,20 While amiodarone has been shown to enhance the anticoagulant effects of warfarin, resulting in an increased haemorrhagic risk,21-23 no dronedarone-warfarin interactions have been noted. Dronedarone has also been shown to have rhythm- and rate-controlling effects in randomised controlled trials of patients with AF or AFL.

In the US-based American-Australian-African Trial with Dronedarone in Atrial Fibrillation/Flutter Patients for the Maintenance of. Sinus Rhythym (ADONIS) study, the median time to recurrence of AF or AFL increased significantly (x2.67) from 59 days on placebo to 158 days with dronedarone (p=0.002).20 Additionally, the mean ventricular rate at first AF recurrence reduced from 116.6 on placebo to 104.6 with dronedarone (p<0.001). In the European Trial In Atrial Fibrillation or Flutter Patients Receiving Dronedarone for the Maintenance of Sinus Rhythm (EURIDIS) study, the time to a recurrence of AF increased significantly (x2.34) from 41 days on placebo to 96 days on dronedarone (p=0.01), while the mean ventricular rate at first AF recurrence was 117.5 beats per minute on placebo compared with 102.3 beats per minute on dronedarone (p<0.001).20

While dronedarone has shown rhythm- and rate-controlling effects, safety concerns were raised by the randomised controlled Antiarrhythmic Trial with Dronedarone in Moderate-to-Severe Congestive Heart Failure Evaluating Morbidity Decrease (ANDROMEDA) trial.24 This trial accrued patients with recently decompensated HF (but without AF) requiring recent hospitalisation or referral to a specialised HF clinic for worsening symptoms. At a median follow-up of two months, there were significantly more deaths in the dronedarone treatment group versus the placebotreated group (25 versus 12, respectively; p=0.03). These observations led to the trial being prematurely stopped. The large randomised controlled ATHENA trial evaluated the efficacy of dronedarone versus placebo, on top of standard treatment, in preventing cardiovascular hospitalisation or death.25,26 The trial accrued 4,628 patients with a recent history of AF/AFL who were in sinus rhythm or who were to be converted to sinus rhythm. In addition, patients were either ÔëÑ75 years of age or >70 years of age with at least one risk factor for death. At a mean follow-up of 21±5 months, there was a significant reduction in the time to first event of cardiovascular hospitalisation or death in the dronedarone-treated group versus the placebo-treated group (31.9 versus 39.4%, respectively; p<0.001). The reduction in cardiovascular hospitalisations and mortality in the dronedarone group was driven by a subtantial reduction of AF-related hopitalisations, but other causes for CV admissions were also reduced. These data are consistent with a post hoc analysis from the ADONIS and EURIDIS trials, which accrued patients with AF or AFL. The results of this analysis showed that there were fewer hospitalisations due to cardiovascular causes or deaths in the dronedarone-treated group compared with the placebo-treated group (22.8 versus 30.9%, respectively; p=0.01).20 Furthermore, a post hoc analysis of the pivotal ATHENA study has shown dronedarone to significantly reduce the risk of stroke in patients with AF. The risk reduction was from 1.8 to 1.2% per year (p=0.027). This is a novel finding as there is no previous evidence linking AADs to a reduction in the risk of stroke in AF patients. These data, showing improvements in clinical outcomes suggest that novel multichannel-blocking AADs are still very relevant in the treatment of AF. The atrial-selective strategy is still promising, but it should be noted that the expression of atrial ionic channels is different in the re-modelled atrium versus in the normal healthy atria; consequently, atrial-specific drugs may have reduced efficacy.27,28

Influence of Rate versus Rhythm Control Trials on Antiarrhythmic Drug Development

The relative benefits of a rate-controlling strategy versus a rhythmcontrolling strategy is a subject of much debate, especially as several key trials failed to show survival benefits for rhythm control strategies for AF compared with rate control. The Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM),29 Pharmacologic Intervention in Atrial Fibrillation (PIAF),30 Rate Control versus Electrical Cardioversion for Persistent Atrial Fibrillation Study (RACE),31 Strategies of Treatment of Atrial Fibrillation (STAF)32 and How to Treat Chronic Atrial Fibrillation (HOT CAFE)33 trials showed no significant difference in mortality, major bleeding and thrombo-embolic events between rhythm control and rate control strategies in patients with AF. Notably, the pivotal AFFIRM study, which enrolled just over 4,000 AF patients with a high risk of stroke or death, showed the incidence of all-cause death to be similar in the rhythm control versus rate control groups (23.8 versus 21.3%, respectively; p=0.08).29 The AFFIRM investigator remarked that management of AF with the rate control strategy may offer potential advantages such as a lower risk of adverse drug effects, including bradycardia, gastrointestinal events and pulmonary events. A meta-analysis of these five rate versus rhythm control trials have shown no significant difference in all-cause mortality or in ischaemic stroke incidence between the two groups (p=0.09 and p=0.30, respectively).34 The recent AF-CHF trial evaluated the rhythm versus rate control strategies in congestive HF patients who had left ventricular ejection fraction Ôëñ35% and a documented recent episode of atrial fibrillation.35 Over a treatment period of 60 months, there was no significant difference in the survival rate (% death from cardiovascular causes) of the patients in the two groups (see Figure 2). The results of these recent rate versus rhythm control trials have the potential to influence the development of new AADs, particularly from a regulatory standpoint, with rhythm-controlling drugs coming under increased scrutiny. Novel agents will need to demonstrate both effective suppression of AF symptomology and improved side effect profiles. While rhythm-controlling drugs have proven benefits, especially in terms of symptomatic relief in AF, a rate control plus anticoagulation strategy may be perceived to be a more suitable option for many patients. However, the development of AADs should continue to focus on both strategies.

Outcome Measures for New Antiarrhythmic Drug Trials

The outcome measures used in trials of new AADs should be able to demonstrate the ability of a drug to prevent the burden of AF-related morbidity and mortality and be associated with a good safety profile. The choice of ideal outcome measures in AAD trials should be largely based on the requirements of the regulatory agencies; such measures would be able to demonstrate a drug's safety and efficacy and their superiority over currently available drugs, thereby facilitating regulatory approval. Members of the FDA's Cardiovascular and Renal Drugs Advisory Committee have indicated that the prevention of AF recurrence is still a valid end-point and that symptom reduction is still desirable.36 However, the ability to prevent AF recurrence alone may be insufficient in the bid to gain approval. Safety is of primary importance; this is where the problem often arises with AADs as these agents have been shown to be associated with severe adverse effects.6,7 Therefore, it is of the utmost important for trials to demonstrate the safety of new AADs. If a trial is further able to demonstrate improvements in clinical outcome with a novel drug and also correlate the prevention of AF recurrence (a surrogate end-point) with improvement in clinical outcomes, the drug will have a higher possibility of gaining regulatory approval. For instance, in the previously mentioned ATHENA trial, dronedarone use prolonged the time to a first event of cardiovascular hospitalisation or death; this duration was positively correlated with the prevention of arrhythmia. This study also identified a subset of patients with AF in whom such outcome data (time to first event of cardiovascular hospitalisation or death) may be very relevant. Based on this, it is likely that future trials of AADs will use the same hard end-points as the ATHENA study to evaluate the efficacy and safety of the drugs in different populations. This would provide an opportunity to identify the patient population that would benefit the most from the drug, which would then have a good chance of gaining approval for that specific patient population.

Issues in Pharmacological Cardioversion

Cardioversion of AF is the process of restoring sinus rhythm, and often involves electrical cardioversion (direct-current cardioversion). Electrical cardioversion has shown a high acute success rate (90%) in selected patients.37 Nevertheless, it is an expensive and complex procedure, often requiring specialists to carry out general anaesthesia or sedation during the procedure. The alternative is pharmacological cardioversion with AADs. There are few controlled studies comparing pharmacological cardioversion with direct-current cardioversion. One of the few published studies showed that the efficacy of the initial attempt for cardioversion was similar with pharmacological or electrical cardioversion.38 However, anecdotally, many clinicians prefer having the pharmacological option, especially for rapid conversion. Additionally, the strategy of pre-treatment with an AAD before direct-current cardioversion has been demonstrated to be more beneficial than electrical cardioversion alone. The combination therapy strategy may be more effective in lowering the threshold of cardioversion, in restoring normal sinus rhythm and in preventing the recurrence of AF versus electrical cardioversion alone.2 New AADs that offer a short duration of observation after infusion for cardioversion would also be preferable.

The Role of Antiarrhythmic Drugs in a World of Curative Ablation Therapy

Atrioventricular (AV) nodal ablation, coupled with pacing, is a highly effective non-pharmacological option for rate control in select patients,39 but is typically recommended as second-line treatment after drug therapy. Similarly, left atrial (LA) ablation for rhythm control is second-line treatment after AADs.2 Catheter-directed ablation techniques and strategies have evolved in recent years to include pulmonary vein isolation (PVI) and linear ablation.40-42 However, there are limited available studies for catheter ablation strategies.2 The predicted prevalence of AF in the US is expected to be as high as nearly 16 million by 2050 (see Figure 3).43 Such a high prevalence would present a substantial increase in the demand for these procedures as they are already becoming more commonplace. However, this is problematic as ablation procedures can only becarried out at specialised centres by skilled professionals.

The current lack of scientific data demonstrating long-term efficacy ofablation in preventing AF suggests that there is still very much a need to develop novel AADs. Pharmacological therapy provides options for multiple indications, including elderly patients and those who are failed or have undergone a partially effective ablation, as well as those for whom AF ablation is not accessible. Additionally, AADs may be more cost-effective as the annual cost of medical therapy has been shown to be lower than catheter ablation in patients with permanent AF (US$4,176-5,060 versus US$16,278-21,294, respectively).44 The use of ablation is now being extensively surveyed and data will become available providing information on the rate of AF recurrence associated with ablation techniques. Hybrid therapeutic strategies involving pharmacotherapy with AADs plus ablation may be an attractive option as it could allow effective rate control and prevention of AF recurrence, and may also reduce the economic burden of disease management, particularly due to a reduction in the need for ablations. Such a strategy is a key area for further research.

Conclusions

Regulatory considerations for new drug applications for new AADs may include the expectation of a positive placebo-controlled clinical outcome trial similar to ATHENA. If this occurs, it is possible that sponsors of novel AF therapies may be dissuaded from continuing drug development. However, if clinical outcome is not required, agencies may still require evidence of unequivocal safety; this would require enrolment of thousands of patients in an expensive and lengthy non-inferiority trial. Another potential hurdle could be that performing placebo-controlled clinical outcome trials, as was the case with ATHENA, may be deemed unethical, and regulatory agencies may instead insist on a positive control (i.e. dronedarone) trial. Thus, unknown regulatory challenges may present obstacles in the development of promising novel therapies for AF. If the bar is sufficiently raised and includes the expectation of a positive placebocontrolled clinical outcome trial similar to that of ATHENA, it is possible that sponsors of novel AF therapies may be dissuaded from continuing drug development. However, if clinical outcome is not required, agencies may still require evidence of unequivocal safety; this would require enrolment of thousands of patients in an expensive and lengthy non-inferiority trial. Another potential hurdle could be that performing placebo-controlled clinical outcome trials, as took place in the ATHENA study, may be deemed unethical, and regulatory agencies may instead insist on a positive control (i.e. dronedarone) trial. Thus, unknown regulatory challenges may present obstacles in the development of promising novel therapies for AF.

The current strategies for the development of new AADs include drugs with atrial-selective or novel non-selective multichannelblocking mechanisms. In the future, atrial-selective drugs will need to demonstrate equivalent or superior safety and efficacy to the non-selective multichannel-blocking approach, especially in light of the approval of dronedarone, a novel multichannel-blocking drug. There is also the question of whether there is a need to continue to develop new AADs in the age of direct-current cardioconversion, potential curative procedures with ablation and the results of the major rate versus rhythm trials. An immediate impact is that there will be an increased impetus on the demonstration of safety and efficacy of new agents over current options. Desirable end-points for future clinical trials of new AADs include symptom reduction, correlation of AF recurrence prevention with improvement in clinical outcomes and efficacy and safety in different patient populations. Novel pharmacological agents with improved safety profiles and demonstrated efficacy will continue to have a place in the management of AF. Pharmacotherapy offers more choices for both clinicians and patients, both as primary therapy and as adjunctive or complementary options in clinical settings.

References

  1. Go AS, Hylek EM, Phillips KA, et al., Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study, JAMA, 2001;285:2370-75.
    Crossref | PubMed
  2. Fuster V, Ryden LE, Cannom DS, et al., ACC/AHA/ESC 2006 Guidelines for the Management of Patients with Atrial Fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Revise the 2001 Guidelines for the Management of Patients With Atrial Fibrillation): developed in collaboration with the European Heart Rhythm Association and the Heart Rhythm Society, Circulation, 2006;114:e257-354.
    Crossref | PubMed
  3. Benjamin EJ, Wolf PA, D'Agostino RB, et al., Impact of atrial fibrillation on the risk of death: the Framingham Heart Study, Circulation, 1998;98:946-52.
    Crossref | PubMed
  4. Ringborg A, Nieuwlaat R, Lindgren P, et al., Costs of atrial fibrillation in five European countries: results from the Euro Heart Survey on atrial fibrillation, Europace, 2008;10: 403-11.
    Crossref | PubMed
  5. Vaughan Williams EM, A classification of antiarrhythmic actions reassessed after a decade of new drugs, J Clin Pharmacol, 1984;24:129-47.
    Crossref | PubMed
  6. Camm AJ, Safety considerations in the pharmacological management of atrial fibrillation, Int J Cardiol, 2008;127: 299-306.
    Crossref | PubMed
  7. Naccarelli GV, Wolbrette DL, Khan M, et al., Old and new antiarrhythmic drugs for converting and maintaining sinus rhythm in atrial fibrillation: comparative efficacy and results of trials, Am J Cardiol, 2003;91:15-26D.
    Crossref | PubMed
  8. Ehrlich JR, Biliczki P, Hohnloser SH, et al., Atrial-selective approaches for the treatment of atrial fibrillation, J Am Coll Cardiol, 2008;51:787-92.
    Crossref | PubMed
  9. Fedida D, Orth PM, Chen JY, et al., The mechanism of atrial antiarrhythmic action of RSD1235, J Cardiovasc Electrophysiol, 2005;16:1227-38.
    Crossref | PubMed
  10. Dorian P, Pinter A, Mangat I, et al., The effect of vernakalant (RSD1235), an investigational antiarrhythmic agent, on atrial electrophysiology in humans, J Cardiovasc Pharmacol, 2007;50:35-40.
    Crossref | PubMed
  11. Roy D, Pratt CM, Torp-Pedersen C, et al., Vernakalant hydrochloride for rapid conversion of atrial fibrillation: a phase 3, randomized, placebo-controlled trial, Circulation, 2008;117:1518-25.
    Crossref | PubMed
  12. Kowey PR, Roy D, Pratt CM, Efficacy and safety of vernakalant hydrochloride injection for the treatment of atrial fibrillation after valvular or coronary artery bypass surgery, Circulation, 2007;116:7.
  13. Stiell I, Roy D, Pratt C, et al., RSD1235 effectively converts acute atrial fibrillation to sinus rhythm independent of background use of oral rateor rhythmcontrol medications (abstract 148), Ann Emerg Med, 2006;48:S46.
  14. Astellas Pharma US Inc, Briefing Materials for the Cardiovascular and Renal Drugs Advisory Committee, 2007. Available at: www.fda.gov/ohrms/dockets/ac/07/ briefing/2007-4327b1-01-astellas-backgrounder.pdf
  15. Roy D, Rowe BH, Stiell IG, et al., A randomized, controlled trial of RSD1235, a novel anti-arrhythmic agent, in the treatment of recent onset atrial fibrillation, J Am Coll Cardiol, 2004;44:2355-61.
    Crossref | PubMed
  16. de Haan S, Greiser M, Harks E, et al., AVE0118, blocker of the transient outward current (I(to)) and ultrarapid delayed rectifier current (I(Kur)), fully restores atrial contractility after cardioversion of atrial fibrillation in the goat, Circulation, 2006;114:1234-42.
    Crossref | PubMed
  17. Schotten U, de Haan S, Verheule S, et al., Blockade of atrial-specific K+-currents increases atrial but not ventricular contractility by enhancing reverse mode Na+/Ca2+-exchange, Cardiovasc Res, 2007;73:37-47.
    Crossref | PubMed
  18. Wegener FT, Ehrlich JR, Hohnloser SH, Dronedarone: an emerging agent with rhythm- and rate-controlling effects, J Cardiovasc Electrophysiol, 2006;17(Suppl. 2):S17-20.
    Crossref | PubMed
  19. Zareba KM, Dronedarone: a new antiarrhythmic agent, Drugs Today (Barc), 2006;42:75-86.
    Crossref | PubMed
  20. Singh BN, Connolly SJ, Crijns HJ, et al., Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter, N Engl J Med, 2007;357:987-99.
    Crossref | PubMed
  21. Muller FO, Steyn JM, Hundt HK, et al., Warfarin bio-availability. A comparison of 4 products, S Afr Med J, 1988;74:566-7.
    PubMed
  22. Breckenridge A, Orme M, Kinetics of warfarin absorption in man, Clin Pharmacol Ther, 1973;14:955-61.
    Crossref | PubMed
  23. Naccarelli GV, Rinkenberger RL, Dougherty AH, et al., Amiodarone: pharmacology and antiarrhythmic and adverse effects, Pharmacotherapy, 1985;5:298-313.
    Crossref | PubMed
  24. Kober L, Torp-Pedersen C, McMurray JJ, et al., Increased mortality after dronedarone therapy for severe heart failure, N Engl J Med, 2008;358:2678-87.
    Crossref | PubMed
  25. Hohnloser SH, Connolly SJ, Crijns HJ, et al., Rationale and design of ATHENA: A placebo-controlled, double-blind, parallel arm Trial to assess the efficacy of dronedarone 400 mg bid for the prevention of cardiovascular Hospitalization or death from any cause in patiENts with Atrial fibrillation/atrial flutter, J Cardiovasc Electrophysiol, 2008;19:69-73.
    Crossref | PubMed
  26. Hohnloser SH, Crijns HJ, van Eickels M, et al., Effect of dronedarone on cardiovascular events in atrial fibrillation, N Engl J Med, 2009;360:668-78.
    Crossref | PubMed
  27. Schotten U, Duytschaever M, Ausma J, et al., Electrical and contractile remodeling during the first days of atrial fibrillation go hand in hand, Circulation, 2003;107:1433-9.
    Crossref | PubMed
  28. Van Wagoner DR, Pond AL, McCarthy PM, et al., Outward K+ current densities and Kv1.5 expression are reduced in chronic human atrial fibrillation, Circ Res, 1997;80:772-81.
    Crossref | PubMed
  29. Wyse DG, Waldo AL, DiMarco JP, et al., A comparison of rate control and rhythm control in patients with atrial fibrillation, N Engl J Med, 2002;347:1825-33.
    Crossref | PubMed
  30. Hohnloser SH, Kuck KH, Lilienthal J, Rhythm or rate control in atrial fibrillation-Pharmacological Intervention in Atrial Fibrillation (PIAF): a randomised trial, Lancet, 2000;356:1789-94.
    Crossref | PubMed
  31. Van Gelder IC, Hagens VE, Bosker HA, et al., A comparison of rate control and rhythm control in patients with recurrent persistent atrial fibrillation, N Engl J Med, 2002;347:1834-40.
    Crossref | PubMed
  32. Carlsson J, Miketic S, Windeler J, et al., Randomized trial of rate-control versus rhythm-control in persistent atrial fibrillation: the Strategies of Treatment of Atrial Fibrillation (STAF) study, J Am Coll Cardiol, 2003;41:1690-96.
    Crossref | PubMed
  33. Opolski G, Torbicki A, Kosior DA, et al., Rate control versus rhythm control in patients with nonvalvular persistent atrial fibrillation: the results of the Polish How to Treat Chronic Atrial Fibrillation (HOT CAFE) Study, Chest, 2004;126:476-86.
    Crossref | PubMed
  34. de Denus S, Sanoski CA, Carlsson J, et al., Rate versus rhythm control in patients with atrial fibrillation: a meta-analysis, Arch Intern Med, 2005;165:258-62.
    Crossref | PubMed
  35. Roy D, Talajic M, Nattel S, et al., Rhythm control versus rate control for atrial fibrillation and heart failure, N Engl J Med, 2008;358:2667-77.
    Crossref | PubMed
  36. Hiatt WR, Lincoff AM, Harrington RA, Acute pharmacological conversion of atrial fibrillation to sinus rhythm: is short-term symptomatic therapy worth it? A report from the December 2007 Meeting of the Cardiovascular and Renal Drugs Advisory Committee of the Food and Drug Administration, Circulation, 2008;117: 2956-7.
    Crossref | PubMed
  37. Lip GY, Tse HF, Management of atrial fibrillation, Lancet, 2007;370:604-18.
    Crossref | PubMed
  38. de Paola AA, Figueiredo E, Sesso R, et al., Effectiveness and costs of chemical versus electrical cardioversion of atrial fibrillation, Int J Cardiol, 2003;88:157-66.
    Crossref | PubMed
  39. Wood MA, Brown-Mahoney C, Kay GN, et al., Clinical outcomes after ablation and pacing therapy for atrial fibrillation : a meta-analysis, Circulation, 2000;101:1138-44.
    Crossref | PubMed
  40. Willems S, Klemm H, Rostock T, et al., Substrate modification combined with pulmonary vein isolation improves outcome of catheter ablation in patients with persistent atrial fibrillation: a prospective randomized comparison, Eur Heart J, 2006;27:2871-8.
    Crossref | PubMed
  41. Ouyang F, Bansch D, Ernst S, et al., Complete isolation of left atrium surrounding the pulmonary veins: new insights from the double-Lasso technique in paroxysmal atrial fibrillation, Circulation, 2004;110:2090-96.
    Crossref | PubMed
  42. Jais P, Hocini M, Sanders P, et al., Long-term evaluation of atrial fibrillation ablation guided by noninducibility, Heart Rhythm, 2006;3:140-45.
    Crossref | PubMed
  43. Miyasaka Y, Barnes ME, Gersh BJ, et al., Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence, Circulation, 2006;114: 119-25.
    Crossref | PubMed
  44. Khaykin Y, Morillo CA, Skanes AC, et al., Cost comparison of catheter ablation and medical therapy in atrial fibrillation, J Cardiovasc Electrophysiol, 2007;18:907-13.
    Crossref | PubMed