You are on page 1of 10

European Journal of Internal Medicine 20 (2009) 672–681

Contents lists available at ScienceDirect

European Journal of Internal Medicine


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / e j i m

Review article

Atrial fibrillation: Mechanistic insights and treatment options


Sunil T. Mathew a,⁎, Jigar Patel b, Satheesh Joseph c
a
Cardiovascular Section, University of Oklahoma Health Sciences Center, University of Oklahoma School of Medicine, Oklahoma City, OK, USA
b
Department of Internal Medicine, University of Oklahoma Health Sciences Center, University of Oklahoma School of Medicine, Oklahoma City, OK, USA
c
Brookhaven Heart, 100 Hospital Rd., Suite 120, East Patchogue, NY 11772, USA

a r t i c l e i n f o a b s t r a c t

Article history: Atrial fibrillation (AF) remains the most common clinically encountered arrhythmia. Unlike supraventricular
Received 19 December 2008 arrhythmias that use a defined mechanism, AF involves a wide spectrum of arrhythmias from lone AF to
Received in revised form 23 June 2009 paroxysmal to chronic AF. AF is an arrhythmia that may develop in several ways. Mechanical remodeling
Accepted 22 July 2009
manifests as decreased atrial contractility and increased atrial compliance which leads to a stretch of the
Available online 27 September 2009
atrial myocardium. Atrial remodeling may also increase in atrial fibrosis which can slow conduction velocity
and can shorten the refractory period in atria with long-standing AF. It is still unclear whether initiation of AF
Keywords:
Arrhythmia activates direct inflammatory effects or whether the presence of a pre-existing systemic inflammatory state
Atrial fibrillation promotes further persistence of AF.
Arrhythmia mechanism Currently, the patient population undergoing AF ablation has greatly expanded. Patients are older and have
Arrhythmia treatment larger left atrial size and are more likely to have persistent/permanent AF. It is likely that AF comprises a
spectrum of disease with no single mechanism adequate enough to comprehensively explain AF and its
variability. The management of patients with AF involves elements of anticoagulation, rate control and
rhythm control and such treatment strategies are not necessarily mutually exclusive of each other.
© 2009 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.

1. Introduction possible mechanisms which may be interrelated. This paper reviews


this clinically widespread arrhythmia. Both traditional and more
Atrial fibrillation (AF) is the most common clinically encountered contemporary theories responsible for AF as well as standard and
arrhythmia [1]. The arrhythmia is associated with heart failure, se- evolving treatment modalities are also reviewed herein.
nescence, and numerous diseases related to senescence and has an
incidence of more than 2.3 million in the United States. However, the 2. Basic science insights on atrial fibrillation mechanism(s)
mechanisms underlying AF are only partially understood and recent
epidemiologic data indicate that AF prevalence is not explained by age 2.1. Rotors and wavelets
alone [2]. Furthermore, additional risk markers such as obesity [3,4],
obstructive sleep apnea [5,6], anger and hostility [7], and long-term Several theories attempt to explain different aspects of such
alcohol consumption [8–10] only partially account for the increase in mechanism(s), however a unifying theory remains lacking. Three
AF. Multiple AF descriptors have developed in light of such wide- major theories are often included in this constituency, including
spread prevalence of the arrhythmia. Various descriptors such as lone (a) the random reentry model; (b) the single or multiple rapidly firing
AF, paroxysmal AF, and chronic AF had been previously used some- ectopic foci; and (c) the multiple reentrant model.
what indiscriminately. Recently however a consensus classification
scheme has been proposed to simplify its description with respect to 2.1.1. Random reentry
its clinical relevance (Fig. 1). Moe and Abildskov's hypothesis [11] of random reentry has been
Although significant progress in understanding the mechanism of one of the prevailing concepts. Its basis centers on multiple wandering
this arrhythmia has been accomplished with the advent of catheter atrial reentrant wavelets colliding with each other to either self-
ablation, the pathophysiology of AF is complex and likely has many terminate or produce daughter wavelets which sustain atrial excita-
tion. Theories describing the genesis of AF typically have originated
from animal models describing these random atrial reentry wavelets.
Canine models have demonstrated that these AF pathways are vagally
⁎ Corresponding author. Fax: + 1 866 384 0496. mediated and are determined by a critical amount of atrial refrac-
E-mail address: stmathew@gmail.com (S.T. Mathew). toriness and excitability, not on atrial anatomy [11–14].

0953-6205/$ – see front matter © 2009 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.ejim.2009.07.011
S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681 673

2.2. Modulating factors

Other basic mechanisms include those that modulate arrhythmo-


genesis [22] which often occur in conjunction with those mechanisms
that are thought to initiate or perpetuate AF. These modulation factors
can further be described as potentially modifiable or non-modifiable
(i.e. age or genetics). As discussed below, there are many modifiable
modulating factors that may maintain or inhibit AF. The major
modulating factors such as endocrine activity, pulmonary vein and
atrial function, are typically more familiar to the practicing clinician
and will be described. Those less prevalent clinically, such as mechan-
ical and electrical atrial remodeling and inflammatory processes will
also be reviewed, as well as the advanced concept of AF modulation by
the autonomic nervous system. Wyse and Gersh have proposed
schematics to illustrate the presumed relationship among modulation
factors to AF (Fig. 2) as well as how these relate to a trigger and/or
substrate as a fundamental mechanism(s) thought to initiate and
sustain AF (Fig. 3). Entities thought to be AF “triggers” are more
associated with initiating AF and may be more predominant in the
mechanism of paroxysmal AF. Alternatively, those described as
“substrates” are more likely associated with influencing persistent AF.

2.2.1. Mechanical and electrical atrial remodeling


Fig. 1. Updated atrial fibrillation classification schema. Our understanding of AF was broadened by the recognition that
(Adapted from Estes NA 3rd, Halperin JL, Calkins H, et al. J Am Coll Cardiol. 2008 Feb once AF initiated, atrial electrophysiological properties changed and a
26;51(8):865–84.) lower threshold induced the arrhythmia. This process explained AF
propagation by a process known as electrical remodeling [24]. The
primary stimulus needed to induce electrical remodeling was an
estimated tenfold increase in the atrial rate caused by AF [25]. Similar
2.1.2. Multiple foci changes were also produced by any form of sufficiently rapid atrial
Somewhat in contrast to the random reentry model, another tachycardia [25].
theory suggests single or multiple rapidly firing ectopic foci. This is not The cellular basis for the observed changes from AF mediated
as widely acknowledged, based in part from studies supporting tachycardia is thought to be due to changes in calcium (Ca2+) loading.
verification of the multiple reentrant concept by Allessie et al. [15,16]. Ca2+ enters the cells through inward L-type Ca2+ current (ICa 2+) and
with each action potential, there is a substantial increase in cellular
2.1.3. Multiple reentry Ca2+ loading in the presence of the arrhythmia [26]. The ensuing
Though multiple reentrant theory appeared to best explain adaptive responses that occur decrease Ca2+ loading by reducing ICa2+,
persistent AF [12,14], there still lacks a general inability of these but at the expense of a shorter action potential duration, which
principal theories to fully describe AF and this further underscores increases the propensity for atrial fibrillation [25,26]. These AF in-
both the difficulty in understanding this arrhythmia. duced electrophysiological changes promote a vicious cycle of ar-
rhythmia that enhances the ability of the disorder to sustain itself, and
2.1.4. New data also create an increasing vulnerability to relapse [24,27–29] (often
Contemporary investigations have only continued to add complex- described as “AF begets AF”) [24].
ity. For example, the initiation of AF from anatomic substrates in the
area around the pulmonary veins [17] question the random reentry
hypothesis and have resurrected prior theories such as the single
source of stable reentry. An attempt at understanding the various
putative mechanisms is required in order to adequately utilize some
of the state-of-the-art therapies that are now available.
Jalife and colleagues, using optical mapping studies on an ovine
model, demonstrated that a primary localized generator (or rotor)
comprising either an ectopic focus or single small reentry circuit
[18,19] in the atria is responsible for AF initiation. In this model, a
functional, anatomic reentry occurs with spiral waves rotating around
reentrant circuits of about one centimeter diameter and is thought to
be the likely source of AF [20,21]. They have also shown that these
dominant rotors that drive AF consistently arise from the left atrium
with passive activation of the right atrium [18,19]. While such studies
have shown that in some cases the relative roles of focal reentry and
abnormal automaticity in the pulmonary vein region may be more
relevant, it is still unclear whether significant rotors exist in humans
and what their location may be. There is also ambiguity as to the
differences between the apparently disparate concepts of multiple
Fig. 2. Schematic illustrating the numerous “modulating factors” that influence and may
wavelet reentry and the primary rotor model: some have hypothe- contribute to the pathogenesis of atrial fibrillation.
sized that it is possible that they may have more similarities than (Wyse DG, Gersh BJ. Atrial fibrillation: a perspective: thinking inside and outside the
differences [22,23]. box. Circulation 2004 Jun 29;109(25):3089–95.)
674 S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681

development of smaller sized intra-atrial circuits [30]. The size of the


atrial reentry circuits depends on the circuit's wavelength: those
with a short wavelength (or smaller atrial circuits) will maintain
several reentry circuits and thereby promote the continuation of AF
(Fig. 4) [28,30]. Larger atria are more likely than smaller atria to
promote smaller intra-atrial circuits [30].

2.2.2. Inflammation/fibrosis
Atrial remodeling may also increase in atrial fibrosis. This process can
slow conduction velocity and may change the atrial refractory period
with long-standing AF [31,32]. It is still unclear whether initiation of AF
activates direct inflammatory effects or whether the presence of a pre-
existing systemic inflammatory state promotes further persistence of AF
[33]. One of the first studies comparing inflammatory markers to atrial
remodeling was reported by Frustaci et al. [34]. Nakamura et al. [35]
confirmed this association by showing that atrial biopsies from patients
with lone AF had higher amounts of atrial fibrosis, inflammatory
Fig. 3. Schematic illustrating how modulating factors relate to the various mechanisms
thought to invoke reentry.AF initiation and perpetuation are illustrated as well as how infiltrates, and myocyte necrosis compared to those with sinus rhythm.
AF can produce modulating factors that further perpetuate it or produce manifestations Canine models with sustained AF demonstrated active atrial perimyo-
of AF. carditis with inflammatory infiltrates, lipid degeneration, and fibrosis
(Wyse DG, Gersh BJ. Atrial fibrillation: a perspective: thinking inside and outside the [36]. Furthermore, not only have increased C-reactive protein (CRP)
box. Circulation 2004 Jun 29;109(25):3089–95.)
levels been demonstrated as early as within the first 24-hours of the
onset of AF [37], patients with persistent AF have also shown markedly
Although it is well established that the perpetuation of AF causes increased CRP levels when compared to patients with paroxysmal AF
electrical remodeling, AF's links to contractile and structural remodel- (independent of other risk factors) [38]. This suggests that inflammation
ing remain controversial. Mechanical remodeling is thought to com- may also promote the persistence of AF.
prise both contractile and structural remodeling of the atria as a result Corollaries to these observations came from a positive effect of
of AF. Whereas electrical and contractile atrial remodeling may be corticosteroid treatment in a randomized study of 216 patients un-
reversible after the cessation of AF, structural remodeling is less likely dergoing coronary or valvular heart surgery [39]. The study demon-
to revert [22,30]. Electrical remodeling and constant changes in the strated that a single dose of dexamethasone was associated with a
L-type Ca2+ channels are both thought to be the primary cause for decreased incidence of new-onset AF in the first 3 days post-surgery.
contractile remodeling. Mechanical remodeling manifests as de- Recently, a similar multi-centered randomized study corroborated
creased atrial contractility and increased atrial compliance which this by showing that the use of intravenous hydrocortisone reduced
causes the atrial myocardium to stretch. Such decreased atrial the incidence of AF after cardiac surgery among 241 consecutive
contractility acts as a stimulus for structural remodeling due to the patients without prior AF or flutter [40]. The pleiotropic effects of
remodeled enlarged atria. This abnormal atrium now becomes an HMG-CoA reductase inhibitors have also been shown in studies which
electro-anatomical substrate for AF by allowing more intra-atrial
circuits to potentially perpetuate due to a reduction in wavelength
(shortening of refractoriness and slowing of conduction) and an
increase in non-uniform atrial tissue [30]. Thus, the remodeled atria
with its enlarged chambers act as AF substrate as it promotes the

Fig. 4. Illustration of the potential interplay of electrical, contractile and structural


feedback loops of atrial remodeling on AF. Increased atrial myocardial stretch is
hypothesized to act as a stimulus for structural remodeling of the atria. The resulting Fig. 5. Illustration of the possible role of oxidative stress and inflammation contributing
enlarged atria allow intra-atrial circuits of small size, due to a reduction in wavelength to electrical remodeling in AF. [ICaL — L-type Ca2+ current; APD — action potential
(shortening of refractoriness and slowing of conduction) and increased non-uniform duration; AERP — atrial effective refractory period; CV — conduction velocity.]
tissue anisotropy (zigzag conduction). (Korantzopoulos P, Kolettis T, Siogas K, Goudevenos J. Atrial fibrillation and electrical
(Allessie M, Ausma J, Schotten U. Electrical, contractile and structural remodeling remodeling: the potential role of inflammation and oxidative stress. Med Sci Monit
during atrial fibrillation. Cardiovasc Res 2002 May;54(2):230–46.) 2003;9(9):RA225-9.)
S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681 675

investigated the association between statin use and incidence of AF. a thicker myocardial extension [55]]. Such observations from the area
The most compelling data come from Kumagai et al. [36] which of the pulmonary veins with the passive activation of the right atrium
utilized atorvastatin on AF in a canine pericarditis model. The ator- [18,19] have lessened the dominance of the multiple wavelet
vastatin group had lower C-reactive protein levels, less pronounced hypothesis and renewed interest in concepts of fibrillatory conduction
fibrosis in the atrial myocardium, and a shorter duration of AF. Fibrosis and a dominant rotor as primary drivers of AF [57]. These observations
and inflammation are thought to change the anatomical and have provided further pathophysiological insight and have also
electrophysiological substrate of the atria [28] and it is thought that helped expand therapy options for current cutting edge pulmonary
the interplay of inflammation may not only be a response to AF but vein ablation procedures.
also an integral part of it [41]. Inflammation and fibrosis in AF merits
further study (Fig. 5) [42] since recovery from electrical remodeling 2.2.5. Autonomics
by conventional pharmacotherapy remains inadequate. The anatomy of the sympathetic and parasympathetic innervation of
the sinus and AV nodes is intricate and each component of the autonomic
2.2.3. Endocrine activity nervous system follows a different course [58]. Parasympathetic neurons
Using a multivariable model based on data from the Framingham extend to the heart through the vagus nerves and the ones destined to
Heart Study (with adjustment for age and other AF risk factors), the innervate the sinus node terminate in the pulmonary vein fat pad,
odds ratio of AF with diabetes was 1.4 for men and 1.6 for women [43]. whereas those that predominantly innervate the AV node terminate in the
Kato et al. hypothesized that endocrine mediated disorders such as inferior vena cava–inferior left atrial fat pad [59]]. Parasympathetic
diabetes mellitus may promote the development of atrial fibrosis denervation by surgical dissection of the fat pads does not influence
leading to a predisposition to AF. Using a rat model, they found that sympathetic control of sinus rate and AV-nodal conduction [57]].
structural remodeling of the atrium characterized by diffuse inter- The electrophysiologic properties of atrial cells are moderated
stitial fibrosis is a major substrate for diabetes related AF [44]. Another differently by parasympathetic and sympathetic influences. For
well-known clinical endocrine modulating factor is the association of example, vagal mediated influences favor atrial macroreentry phe-
AF with hyperthyroidism whereby changes in thyroid hormone nomena and sympathetic influences favor abnormal automaticity and
concentration directly affects the initiation or inhibition of AF [22]. triggered activity [60]. In normal hearts, vagal influences predomi-
There is also growing evidence to suggest a role for the renin– nate, which partially explains why the clinical pattern of vagally
angiotensin system (RAS) in the pathogenesis of AF. Inhibition of RAS mediated paroxysmal AF is often found in young males with no
with captopril or candesartan, yields a reduction in atrial tachycardia detectable heart disease. These patients usually have an ECG pattern of
associated electrical remodeling in dogs subjected to rapid atrial atrial flutter alternating with fibrillation [61]. The vagal effect is to shorten
pacing for a relatively short duration. Among dogs subjected to pacing both the action potential and refractory period. This is recognized to occur
for a longer duration (i.e. 1–5 weeks), RAS inhibition did not appear to heterogeneously within the atrial wall, suggesting that such inhomo-
alter atrial remodeling during the relatively early stages of electrical geneity may contribute to a reentry mechanism rather than hyperexcit-
remodeling [45,46]. The role of RAS in atrial remodeling to sustain AF ability [62]]. Conversely, AF mediated by the sympathetic nervous system
in humans is less clear. Angiotensin II infusion in human subjects has is observed in the presence of most any heart disease, of which the first
not been shown to shorten the atrial refractory period [42] and may effect is often to provoke a vagal withdrawal [59]].
not have a significant acute effect on human atrial electrical re- Although studies have demonstrated that the rate of arrhythmo-
modeling. It may, however, facilitate the role of structural remodeling genicity near the pulmonary vein can be greatly accelerated by
and fibrosis of atrial tissue, which suggests a link between RAS and adrenergic stimulation [63], little is known about the impact of base-
remodeling of atrial tissue [42]. This may be consistent with clinical line autonomic tone and the development of new-onset AF in the
data demonstrating the effectiveness of a RAS blockade in preventing general population. Despite epidemiological evidence from the
new-onset or recurrent AF among those with hypertension and left Framingham Heart Study subjects suggesting that baseline autonomic
ventricular hypertrophy, congestive heart failure, and those under- dysregulation reflected by altered heart rate variability (HRV) is
going electrical cardioversion for AF [47,48]. However, more clinical associated with risk of AF, the association did not persist after
and experimental animal studies are needed in order to define the adjusting for potential confounders. Much of the apparent association
role of RAS inhibition in patients with AF. between HRV and AF in the Framingham cohort was believed to be
mediated by traditional risk factors [64].
2.2.4. Pulmonary veins and atrial function Tomita et al. [65] demonstrated a relationship between the time of
Haissaguerre et al. demonstrated that premature depolarizations paroxysmal AF onset and autonomic tone both before and after
originating from the pulmonary veins could contribute to the gen- paroxysmal AF and found that the autonomic nervous system plays an
eration of AF [17]. They showed that the pulmonary vein foci may important role in both the initiation and termination of paroxysmal
initiate AF by originating as a rapid atrial tachycardia. Subsequent AF. They also found that the onset of paroxysmal AF influences the
studies have shown that AF originating from pulmonary vein foci may autonomic tone at the initiation as well as during termination of
then degenerate as a consequence of atrial remodeling which acts to paroxysmal AF [65]. However, the role of autonomic tone in the
maintain AF by promoting multiple reentry circuits [49,50]. Other initiation of paroxysmal atrial flutter has not been reported.
studies have also shown this activity and implicate the pulmonary These epidemiologic and clinical observations between AF and the
veins in both the initiation as well as maintenance of AF [51,52]. autonomic nervous system gain further credibility with basic science
The pulmonary vein-left atrial junction has also become a target associations. For example, the novel Class III antiarrhythmic agent,
for morphological studies in both animal and human hearts in NIP-142, shows a link between the autonomic impact on AF in a canine
describing AF arrhythmogenicity. The multilayer left atrial myocar- model of atrial fibrillation and flutter. As a primary potassium channel
dium extend in a variable arrangement and distance into each PV blocker, NIP-142 prolonged atrial refractoriness and was effective in
resulting in a complex three-dimensional fiber orientation [53,54]]. treating atrial flutter and vagally-induced AF [66]. Such studies are
These pulmonary vein-left atrial muscle sleeves consist of specialized important as they add insight into the autonomic component of the
conduction cells similar to those found in the sinus node [55]. The arrhythmia. This understanding forms the basis for some of the
transition zones of these myocardial extensions were thickest at the emerging therapies aimed at targeting the autonomic nervous system
venoatrial junction (mean 1.1 mm) and became thinner moving for the treatment of refractory AF. Coumel has suggested that in order to
distally [56]. Their thickness is variable with the inferior walls of the confirm vagally derived initiation of AF, the minutes and hours pre-
superior veins. In contrast, the superior walls of the inferior veins have ceding AF onset need to be recorded in order to demonstrate the
676 S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681

relationship between arrhythmia onset and heart rate changes [60]. The for each disease state and 2 points for prior strokes or transient
well-known mechanism of ectopic foci located in the left atrium or the ischemic attacks) [69] (see Tables 2 and 3). Anticoagulation with
pulmonary veins [17] is compatible with vagal or adrenergic profiles. warfarin is recommended for patients with valvular atrial fibrillation
Such ectopic foci may form the arrhythmia trigger and vagally derived or a CHADS2 score ≥ 2 (INR 2–3 and 2.5–3.5 for patients with me-
arrhythmias are often sensitive to focal ablation [67]. chanical valves), unless contraindicated or there is an annual major
bleeding risk exceeds 3% [76]. Aspirin or warfarin may be used when
3. Clinical science and treatment issues the CHADS2 score = 1. Aspirin, 81–325 mg daily, is recommended in
patients with a CHADS2 score of 0, or if warfarin is contraindicated.
Although there has been much insight regarding the pathophysiolo- Current recommendations include continued anticoagulation with
gical triggers of AF, an understanding of its mechanisms is still inadequate warfarin in patients with sufficient thromboembolic risk whether
[56]. This manifests by less than optimal treatment of this arrhythmia they have persistent or paroxysmal atrial fibrillation [74,77].
despite the development of novel catheter-based ablation therapy and Differing patient selection criteria, endpoints and therapeutic inter-
pharmacotherapy. Though these techniques may in time lead to a cure for ventions in many of the above studies limit the applicability of the findings
the disorder in some patients, focal atrial ablation or pulmonary vein to all AF populations. For example, AFFIRM, RACE and HOT-CAFE did not
isolation is not currently universally effective [68]]. Currently, two evaluate a single drug or type of treatment. Another criticism was that
fundamental strategies dominate treatment options: (i) ventricular rate these trials were not comparisons of sinus rhythm and AF, but rather
control; and (ii) restoration of sinus rhythm to obtain rhythm control. comparisons of the strategy of rate control to that of rhythm control where
the success rate of the rhythm-control strategy was relatively poor [78].
3.1. Treatment insights from clinical trials: rate or rhythm control Up to 35% of rhythm-control patients in AFFIRM were actually using rate-
control drugs by the end of the study. In subanalyses, many patients in the
AF management involves three facets: rate control; prevention of rate-control arm were spontaneously in sinus rhythm by the end of the
thromboembolism; and the restoration of sinus rhythm. The data study period. The same number was 10% in STAF and RACE to 35% in
available on the clinical relationship between sinus node dysfunction AFFIRM which suggests that by switching analysis according to the
and AF suggest that the sinoatrial (SA) node is probably passive during patient's actual rhythm, the benefit of sinus rhythm over AF becomes
AF. Diseases often affect both the SA node and atria simultaneously obvious [77]]. This is consistent with population-based studies such as
which can contribute to persistent AF. This is different from instances SOLVD, DIAMOND and CHF-STAT which have long shown the negative
where abnormal SA node dysfunction contributes to AF maintenance prognostic impact of AF on survival [79]. With the exception of the PIAF
[28]. Theoretically, the basis for restoration of normal sinus rhythm was trial, antithrombotic therapy was discontinued in some patients who were
believed to have several benefits over rate control. These potential assigned to the rhythm-control groups. Withholding antithrombotic
advantages include improved cardiac output from the restoration of therapy is one possible explanation for the higher rate of non-
atrial systolic function which yields improvement in quality of life; cardiovascular mortality observed in the rhythm-control arm of AFFIRM.
greater exercise capacity; lower risk of stroke; the possibility of stopping The majority of strokes in RACE occurred while receiving inadequate
anticoagulation with maintenance of sinus rhythm; and the theoretical anticoagulation therapy. In RACE, 21 of the 35 thromboembolic
benefits of potentially reversing atrial structural or electrical remodeling complications occurred under rhythm control. Six patients had the
[28]. Even in light of these potential advantages, it appears that rhythm event after cessation of oral anticoagulant therapy, with five of them in
control is nearly equivocal to rate control for mortality benefit. apparent sinus rhythm. Asymptomatic episodes of AF may add to
Several trials have analyzed the restoration of normal sinus rhythm continued stroke risk in rhythm-control patients who are believed to be
and demonstrated that not only did rhythm control offer no added in persistent sinus rhythm [80]]. Such thromboembolic complications
morbidity and mortality benefit, the quality of life measures did not despite apparent sinus rhythm contribute to the benefits of warfarin and
differ significantly during follow-up. Such trials included RACE (RAte its ability to reduce the risk of death by a third [75,81]].
Control versus Electrical conversion) [69], AFFIRM (Atrial Fibrillation Younger patients who have greater cumulative risk for developing
Follow-up of Rhythm Management) [70], PIAF (Pharmacological sequelae of chamber remodeling were underrepresented in these trials.
Intervention in Atrial Fibrillation) [71], STAF (Strategies of Treatment The mean age was 70 years in AFFIRM and 68 years in the RACE study. In
of Atrial Fibrillation) [72], and HOT-CAFE (How to Treat Chronic Atrial excluding the younger subgroup, the results of these studies may not be
Fibrillation) [73] (the study parameters and key findings are representative for the full spectrum of AF patients. This younger
summarized in Table 1). In summary, these studies showed that a symptomatic subgroup is thought to represent at least one third of all AF
rate-control strategy was at least as effective as rhythm control and patients [82]. With AFFIRM, there was a trend toward a benefit of
that antithrombotic therapy is important even when a rhythm-control rhythm control for patients younger than 65 years which indicated an
strategy is employed. Though an important caveat to this was that the improved outcome in cases of maintained sinus rhythm [70]. In PIAF,
trials were problematic because of the inability to maintain sinus where the mean age was slightly younger (60–61 years), exercise
rhythm in a large percentage of the sinus-rhythm arm suggesting that tolerance was better with a rhythm-control strategy. A follow-up
the antiarrythmics used proved essentially inadequate for the job and analysis of outcomes in AFFIRM [79] showed that the presence of sinus
no superiority of sinus control could be demonstrated. rhythm was associated with reduced mortality (47% reduction, 99% CI
Regardless of how the rate- or rhythm-control strategy is dealt 28–61; p ≤ 0.0001). The benefits of sinus-rhythm maintenance over
with, attention must also be directed to antithrombotic therapy for time have yet to be carefully evaluated [83] and the optimum resting
prevention of thromboembolism [74]]. In a meta-analysis, the Atrial heart rate or heart rate during exercise remains undefined for patients
Fibrillation Investigators [75] reported that warfarin reduced the risk with permanent AF despite retrospective nonrandomized data of
of death by 33% and that warfarin is the only pharmacologic therapy AFFIRM and the AFFIRM-RACE cooperation [84–87].
that has been reported to improve survival in AF.
Many patients with AF who are thought to be low risk for AF 4. Additional management options
associated stroke do not substantially benefit from anticoagulation.
These patients can be reliably identified using the CHADS2 stroke risk While the cornerstone of long-term AF management is usually
stratification scheme (an acronym for Congestive heart failure, anticoagulation with rate and/or rhythm control, there are several
Hypertension, Age ≥ 75 years, Diabetes and prior Stroke/or transient additional treatment options. The following section will touch on
ischemic attack (with S2 to indicate stroke with a weight of 2 points)). many of the major management options available and describe some
Ischemic stroke risk is estimated with the CHADS2 score (one point of the benefits and disadvantages of each.
S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681 677

Table 1
Randomized trials comparing rate versus rhythm control for atrial fibrillation.

Trial N Mean Hypotheses Rate-control Rhythm- Primary endpoint Outcome and key p-value
age regimen control findings
regimen
PIAF (Pharmacological Intervention 252 61 There is equivalency Diltiazem Amiodarone or Elimination of Both groups showed 0.317
in Atrial Fibrillation) between rate-control cardioversion palpitations, dyspnea, similar improvement in
versus rhythm-control shortness of breath approximately 60% of
strategies for AF and dizzy spells patients after 3 weeks of
treatment in both arms.
NSR maintained: 56% of
rhythm-control versus 10%
of rate-control points
STAF (Strategies of Treatment of 200 66 There is equivalency β-blockers; Cardioversion; Composite of: death, No mortality difference NS
Atrial Fibrillation) between rate-control nondihydropyridine or amiodarone; stroke or TIA, between two groups.
versus rhythm-control calcium blockers; sotalol resuscitation from Rhythm points
strategies for AF digoxin; or cardiac arrest, and hospitalized significantly
cardioversion; or peripheral emboli more/for longer periods.
ablation and/or 70% of rhythm-control
pacemaker points successfully
implantation cardioverted, but NSR
maintained in only 23% at
3 years (versus 9% of points
in rate-control group)
AFFIRM (Atrial Fibrillation Follow-up 4060 70 There is noninferiority β-blockers; Amiodarone, Overall or all-cause No significant difference in 0.08
of Rhythm Management) of rate control versus nondihydropyridine disopyramide, mortality all-cause mortality
rhythm control in calcium blockers; flecainide, • Mortality: rhythm
patients with AF digoxin; or moricizine, control, 23.8%; rate
combinations of procainamide, control, 21.3%
these drugs propafenone, • More adverse drug
quinidine, reactions in rhythm-
sotalol, and control points
combinations • 63% of rhythm-control
points in NSR; 80% of
rate-control points
with adequate HR
control at 5 years
RACE (RAte Control versus 522 68 There is noninferiority Digitalis; or Sotalol; or Composite of Composite endpoint reached 0.11
Electrical conversion) of rate control to nondihydropyridine flecainide; or cardiovascular death, in 17.2% of patients in rate-
rhythm control with calcium-channel propafenone; hospitalization for control arm and 22.6% in
regard to morbidity blockers; or a β- or amiodarone heart failure, rhythm-control arm
and mortality blockers, alone or in thromboembolic • 39% of rhythm-control
combination complications, severe points in NSR versus
hemorrhage, 10% in rate-control group
pacemaker • Thromboembolic
implantation, or severe complications more
drug side effects likely in rhythm group
• Pacemaker implantation
more likely in rhythm
group
HOT-CAFE (HOw to Treat Chronic 206 61 There is equivalency β-blockers; Propafenone Composite of No significant differences 0.71
Atrial Fibrillation) between rate-control nondihydropyridine disopyramide, cardiovascular death, between rate- and rhythm-
versus rhythm-control calcium blockers; sotalol, or thromboembolic control points. No
strategies for AF digoxin; or a amiodarone complications or thromboembolic
combination of these intracranial or other complications observed in
drugs; or major hemorrhage points left in AF; 3 ischemic
cardioversion; or strokes reported in rhythm-
ablation control arm (2 due to
cardioversion, 1 to late AF
recurrence)

4.1. Pacing and atrioventricular nodal ablation mized trials of the procedure versus rate-slowing drug therapy did
not show a significant benefit on endpoints of exercise tolerance,
Among AF patients who are refractory to rate-control objectives, symptomatic improvement or improvement of LV function [90–92].
the “ablate and pace” strategy offers symptomatic relief, but is Other small clinical studies have shown a trend toward a benefit of
considered a therapy of last resort because of the potential negative reducing the frequency of AF, using various alternative pacing
effects of long-term right ventricular (RV)-pacing. A meta-analysis algorithms such as right atrial septal pacing; low interatrial septal
of trials of AV-node ablation and pacing demonstrated symptom pacing; and Bachmann's bundle pacing [93–95]. Some evidence
and exercise tolerance improvement and has provided support for additionally suggests a better outcome with biventricular pacing in
this modality [88]. Procedural shortcomings include complication patients with pre-existing LV-dysfunction [96]. However, AV-nodal
rates associated with the pacemaker insertion or the ablation; per- ablation and permanent pacing is only warranted in patients who
manent pacemaker dependence; and a small risk of sudden death have failed rate-control pharmacotherapy and remain highly symp-
in the early post-ablation period [89]. Furthermore, small rando- tomatic from AF.
678 S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681

Table 2 1998, percutaneous procedures have evolved to replicate and


CHADS2 scoring system for AF. improve on surgical maze techniques. The former is done by making
From: Gage, BF, Waterman, AD, Shannon, W, Boechler M, Rich MW, Radford MJ.
Validation of clinical classification schemes for predicting stroke: results from the
continuous endocardial lesions to encircle the pulmonary veins with
National Registry of Atrial Fibrillation. JAMA 2001;285:2864–2870. the goal of contemporary AF ablation to electrically “disconnect” the
PVs from the rest of the atrium by ablating around the origin of the
Risk factor CHADS2 score (points)
veins. These percutaneous procedures are often considered for
Congestive heart failure (EF b 35%) 1
patients who have failed antiarrhythmic drug therapy. The current,
Hypertension (systolic N 160 mm Hg) 1
Age greater than 75 years 1 patient population undergoing AF ablation has expanded significantly.
Diabetes 1 Patients are older and have larger left atrial size and are more likely to
Stroke (prior cerebral ischemia: CVA or TIA) 2 have persistent AF often having been subjected to fewer prior
CHADS2 is an acronym derived from the stated risk factors and the scoring of 2 for prior antiarrhythmic agents [104].
stroke or TIA. The CHADS2 score is calculated by totaling the scores for each risk factor Although PV isolation results in a long-term clinically satisfactory
present. outcome in an estimated 60–85% [95,105–108] of patients with
Abbreviations: CHF, congestive heart failure; EF, ejection fraction; TIA, transient ischemic
paroxysmal AF, it is clearly not the ideal approach in all AF patients
attack.
[98,109]. Histological studies have shown that complex atrial
anatomy has marked variations in atrial wall thickness [110,111].
4.2. Surgical intervention Adjacent structures of the atrial wall may vary in depth by several
millimeters, so applying the same degree of energy at two contiguous
The goal of AF surgical management is to make atrial incisions at spots can result in excessive heating in one spot and non-transmural
critical locations that create barriers to conduction and prevent heating in the other. Consequently, complete conduction block via
persistent AF preserving sinus node and atrial transport functions radiofrequency application may not always be achieved [112].
[71]. The surgical “maze” or “cut-and-sew” technique has evolved Potential complications of catheter ablation for AF include systemic
tremendously since its inception over 25 years ago. The objective is to thromboembolism, PV stenosis, pericardial effusion, cardiac tampo-
ensure that these transmural incisions isolate the pulmonary veins, nade, and phrenic nerve paralysis [113–115]]. Packer et al. [116] have
while also connecting these incisions to additional surgical incisions suggested that although the short-term safety of newer ablation
around the mitral valve annulus, in order to create electrical barriers techniques has improved, several serious complications remain a
within the atria to prevent macroentrant rhythms—atrial flutter or AF— reality with long-term safety and ablation efficacy rates are still not
from becoming persistent [97]. Success rates [98] from 70% to nearly 95% clearly defined. Finally, unavoidable procedure-related complications
over 15 years of follow-up have been reported in patients undergoing may occur and general skepticism tends to arise which further
mitral valve surgery [99]. Despite its reasonably high success rate, the highlight the need for the development of less complex therapies.
surgical maze is not routinely performed other than for patients
undergoing cardiac surgery because of the requirement of cardiopul- 4.4. Targeting ganglionated plexi
monary bypass. Several less invasive variations are under investigation,
including thoracoscopic and catheter-based epicardial techniques [93]. Coumel first described adrenergically and vagally derived AF in
If the efficacy and safety of these surgical adaptations approach that of patients [60]. Patterson et al. noted that combined parasympathetic
the percutaneous ablative techniques, they may become acceptable and sympathetic nerve stimulation modulate firing in canine
alternatives for a larger proportion of patients with AF. pulmonary veins [117]. The human heart contains a variety of
morphologically distinct nerve terminals concentrated at the gang-
4.3. Percutaneous intervention lionated plexi (GP). They are distributed more widely than has been
described in experimental animals [118]. Other similar arrhythmo-
In most AF patients (94%), the AF focus was in ≥1 of the pulmonary genic foci which may trigger AF have been found in the superior vena
veins (PVs) which was a landmark observation by Haissaguerre et al. cava, the right and left atria, the coronary sinus, and the ligament of
[17]. Extra-PV sites may trigger AF, but is thought to occur in only a Marshall [50]. Focal atrial ablation has a low success rate in patients
minority of patients: no more than 6% to 10% [46]. This facilitated with persistent AF and particularly in patients with the vagotonic type
recognition of the pathophysiological role played by these “muscle of paroxysmal AF. An approach that involves ablation within the left
sleeves”; or extensions of myocardium within the PV in the initiation atrium is likely to yield better results in these subgroups, as well as
of AF. Dominant rotors of AF are localized primarily in the junction those subgroups with paroxysmal AF that do not respond to focal
between the left atrium (LA) and PVs, have been demonstrated by atrial ablation [81,102]. Multivariate analysis indicated that there
several subsequent investigations [77,100–102]. A recent study has were only two independent predictors of recurrent AF after
also demonstrated that the PV-LA region has heterogeneous electro- pulmonary vein isolation: persistent AF at the time of pulmonary
physiological properties capable of sustaining reentry (micro or vein isolation and the vagotonic variety of paroxysmal AF. These
macro) [103]. Since PV isolation was first described for treating AF in results tend to show that the pulmonary veins play a less important
role in the generation of persistent/permanent AF and in the initiation
or maintenance of vagotonic paroxysmal AF than in paroxysmal AF
Table 3 that is either random or adrenergic in onset [107]. Little is known
Risk of stroke by CHADS2 score. about the impact of baseline autonomic tone and the development of
From: Gage, BF, Waterman, AD, Shannon, W, Boechler M, Rich MW, Radford MJ.
new-onset AF in a population-based cohort. Inferences from recent
Validation of clinical classification schemes for predicting stroke: results from the
National Registry of Atrial Fibrillation. JAMA 2001;285:2864–2870. reports have been that stimulation of the GP situated at the PV-atrial
junctions can convert PV focal firing into AF [119] or initiate
CHADS2 score Adjusted stroke rate (per 100 person-years)
paroxysmal AF and can provide a basis for potential therapy [120].
0 1.9
Radiofrequency ablation of the GP can also prevent AF inducibility
1 2.8
2 4.0 [121]. The role of the intrinsic cardiac autonomic nervous system in
3 5.9 the genesis and perpetuation of AF was further emphasized by several
4 8.5 recent clinical reports. This approach centered on ablation of the GP
5 12.5 located in proximity to the PV-atrial junction and resulted in a high
6 18.2
success rate in eliminating symptomatic AF of the vagotonic variety
S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681 679

[122–124]. The involvement of local cardiac autonomic elements electrophysiological instrumentation may contribute to better insight
requires further investigation and it remains to be determined into AF mechanisms for purposes of more effective therapies.
whether targeting the GPs clinically will result in significantly higher
AF ablation success rates. 6. Learning points

4.5. Pharmacologic therapy • Significant progress in understanding the mechanism of this


arrhythmia has been accomplished with the advent of catheter
Pharmacotherapy remains an essential component in the manage- ablation, the pathophysiology of AF is complex and likely has many
ment of AF since invasive procedures are currently not globally possible mechanisms which may be interrelated. This paper reviews
efficacious nor can they be performed on all patients with AF. this clinically widespread arrhythmia. Both traditional and more
Available antiarrhythmic drugs are only modestly effective in contemporary theories responsible for AF as well as standard and
suppressing AF. Drug therapy leading to sinus-rhythm maintenance evolving treatment modalities are also reviewed herein.
may be a marker of improved outcome or less serious disease. Or it
may indicate that, while the ability of treatment to maintain sinus
References
rhythm is inherently beneficial, such benefit may come at the expense
of toxic effects and poor efficacy [27]. It is uncommon for patients to
[1] Go AS, Hylek EM, Phillips KA, Chang Y, Henault LE, Selby JV, et al. Prevalence of
have complete suppression of AF. Successful therapy is often gauged diagnosed atrial fibrillation in adults: national implications for rhythm manage-
by reduction in frequency severity and duration of episodes. ment and stroke prevention: the AnTicoagulation and Risk Factors in Atrial
Most antiarrhythmic drugs used for AF were never designed to Fibrillation (ATRIA) Study. JAMA 2001;285:2370–5.
[2] Chen LY, Shen WK. Epidemiology of atrial fibrillation: a current perspective.
target the atria but rather developed for ventricular arrhythmias Heart Rhythm 2007;4:S1–6.
which partially explains their toxicity limitations predominantly [3] Miyasaka Y, Barnes ME, Gersh BJ, Cha SS, Bailey KR, Abhayaratna WP, et al. Secular
related to prolonged QT. Recently, a meta-analysis of over 11,000 trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to
2000, and implications on the projections for future prevalence. Circulation
patients reported that Class IA, IC, and III drugs are effective in 2006;114:119–25.
maintaining sinus rhythm but also reported an increase in adverse [4] Wang TJ, Parise H, Levy D, D'Agostino Sr RB, Wolf PA, Vasan RS, et al. Obesity and
effects, as well as an increase in mortality with Class IA drugs [125]. the risk of new-onset atrial fibrillation. JAMA 2004;292:2471–7.
[5] Gami AS, Pressman G, Caples SM, Kanagala R, Gard JJ, Davison DE, et al. Association
Specifically, several Class IA (disopyramide phosphate, quinidine of atrial fibrillation and obstructive sleep apnea. Circulation 2004;110:364–7.
sulfate), Class IC (flecainide acetate, propafenone hydrochloride), and [6] Kanagala R, Murali NS, Friedman PA, Ammash NM, Gersh BJ, Ballman KV, et al.
Class III (amiodarone, dofetilide, sotalol hydrochloride) drugs sig- Obstructive sleep apnea and the recurrence of atrial fibrillation. Circulation
2003;107:2589–94.
nificantly reduced recurrence of atrial fibrillation, but they also had
[7] Eaker ED, Sullivan LM, Kelly-Hayes M, D'Agostino Sr RB, Benjamin EJ. Anger and
associated increased withdrawals due to adverse effects. Pooled hostility predict the development of atrial fibrillation in men in the Framingham
analysis of Class IA drugs were associated with increased mortality Offspring Study. Circulation 2004;109:1267–71.
compared with controls and with the exception of amiodarone and [8] Denison H, Jern S, Jagenburg R, Wendestam C, Wallerstedt S. Influence of
increased adrenergic activity and magnesium depletion on cardiac rhythm in
propafenone, each showed an increased propensity toward arrhythmia alcohol withdrawal. Br Heart J 1994;72:554–60.
[125]. Investigational drugs with potentially improved efficacy and [9] Maki T, Toivonen L, Koskinen P, Naveri H, Harkonen M, Leinonen H. Effect of
safety profiles are under development. Some of these novel agents are ethanol drinking, hangover, and exercise on adrenergic activity and heart rate
variability in patients with a history of alcohol-induced atrial fibrillation. Am J
non-selective ion channel blocking drugs or analogs to amiodarone. Cardiol 1998;82:317–22.
Other such experimental drugs are designed to target specific ion [10] Mukamal KJ, Tolstrup JS, Friberg J, Jensen G, Gronbaek M. Alcohol consumption
channels to provide atrial antiarrhythmic effects without ventricular and risk of atrial fibrillation in men and women: the Copenhagen City Heart
Study. Circulation 2005;112:1736–42.
arrhythmias by preferential action on the atria (inhibition of the ITO [11] Moe GK, Abildskov JA. Atrial fibrillation as a self-sustaining arrhythmia
channel) or via selective action on the atria (inhibition of IKUR currents) independent of focal discharge. Am Heart J 1959;58:59–70.
[126]. Combined pharmacologic and nonpharmacologic strategies may [12] Waldo AL. Mechanisms of atrial fibrillation. J Cardiovasc Electrophysiol 2003;14:
S267–74.
ultimately replace current approaches to the treatment of AF. [13] Moe GK, Rheinboldt WC, Abildskov JA. A computer model of atrial fibrillation. Am
Heart J 1964;67:200–20.
5. Conclusion [14] Moe GK. A conceptual model of atrial fibrillation. J Electrocardiol 1968;1:
145–6.
[15] Allessie MALW, Bonke FIM, Hollen J. Experimental evaluation of Moe's multiple
Unlike other supraventricular arrhythmias, AF is more complex. AF is wavelet hypothesis of atrial fibrillation. In: Zipes DPJJ, editor. Cardiovascular
an arrhythmia that may develop in several ways but our basic insight electrophysiology and arrhythmias. Orlando: Grune & Stratton; 1985. p. 265–76.
into its mechanism remains too limited to allow reliable classification. [16] Allessie MALW, Smeets J, Bonke F, Hollen. Total mapping of atrial excitation
during acetylcholine-induced atrial flutter and fibrillation in the isolated canine
Mechanisms operating in AF derived from different animal and heart. Molndal: AB Hassle; 1982.
experimental models are often different in genesis and perpetuation [17] Haissaguerre M, Jais P, Shah DC, Takahashi A, Hocini M, Quiniou G, et al.
from clinical models involving humans [127]. It is likely that AF Spontaneous initiation of atrial fibrillation by ectopic beats originating in the
pulmonary veins. N Engl J Med 1998;339:659–66.
comprises a spectrum of diseases with no single mechanism adequate [18] Mansour M, Mandapati R, Berenfeld O, Chen J, Samie FH, Jalife J. Left-to-right
enough to comprehensively explain the variability of AF. A unifying gradient of atrial frequencies during acute atrial fibrillation in the isolated sheep
conclusion may be that the AF mechanism is multifactorial and we heart. Circulation 2001;103:2631–6.
[19] Mandapati R, Skanes A, Chen J, Berenfeld O, Jalife J. Stable microreentrant sources
cannot be certain about such mechanism of AF in most individual as a mechanism of atrial fibrillation in the isolated sheep heart. Circulation
patients. Despite the treatment implications evidenced in clinical trials, 2000;101:194–9.
it would be an oversimplification to assume that rate control should be [20] Fynn SP, Kalman JM. Pulmonary veins: anatomy, electrophysiology, tachycardia,
and fibrillation. Pacing Clin Electrophysiol 2004;27:1547–59.
the management approach for all patients. The management of patients [21] Jalife J, Berenfeld O, Mansour M. Mother rotors and fibrillatory conduction: a
with AF involves elements of anticoagulation, rate control and rhythm mechanism of atrial fibrillation. Cardiovasc Res 2002;54:204–16.
control and such treatment strategies are not necessarily mutually [22] Wyse DG, Gersh BJ. Atrial fibrillation: a perspective: thinking inside and outside
the box. Circulation 2004;109:3089–95.
exclusive of each other. The development of novel drugs allows for
[23] Cosio FG, Castillo E. Left atrial anatomic remodeling in atrial fibrillation.
improved efficacy, atrial selectivity and reduced toxicity. Evolving J Cardiovasc Electrophysiol 2007;18:53–4.
technological improvement in ablative procedural devices and the [24] Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA. Atrial fibrillation begets atrial
incorporation of real time three-dimensional imaging modalities may fibrillation. A study in awake chronically instrumented goats. Circulation 1995;92:
1954–68.
also improve chances of the termination long-term arrhythmia [25] Wijffels MC, Kirchhof CJ, Dorland R, Power J, Allessie MA. Electrical remodeling
[14,27,126]. It is hoped that continued research and improved due to atrial fibrillation in chronically instrumented conscious goats: roles of
680 S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681

neurohumoral changes, ischemia, atrial stretch, and high rate of electrical [57] Jalife J. Rotors and spiral waves in atrial fibrillation. J Cardiovasc Electrophysiol
activation. Circulation 1997;96:3710–20. 2003;14:776–80.
[26] Sun H, Chartier D, Leblanc N, Nattel S. Intracellular calcium changes and [58] Wu RC, Berger R, Calkins H. Catheter ablation of atrial flutter and macroreentrant
tachycardia-induced contractile dysfunction in canine atrial myocytes. Cardio- atrial tachycardia. Curr Opin Cardiol 2002;17:58–64.
vasc Res 2001;49:751–61. [59] Randall WCAJ. Cardiac electrophysiology: from cell to bedside. In: Zipes DPJJ,
[27] Nattel S, Opie LH. Controversies in atrial fibrillation. Lancet 2006;367:262–72. editor. Nervous control of the heart: anatomy and pathophysiology. Philadel-
[28] Nattel S. New ideas about atrial fibrillation 50 years on. Nature 2002;415:219–26. phia: WB Saunders, Co; 1990. p. 291–9.
[29] Yue L, Feng J, Gaspo R, Li GR, Wang Z, Nattel S. Ionic remodeling underlying [60] Coumel P. Paroxysmal atrial fibrillation: a disorder of autonomic tone? Eur Heart
action potential changes in a canine model of atrial fibrillation. Circ Res 1997;81: J 1994;15(Suppl A):9–16.
512–25. [61] Coumel P. Autonomic influences in atrial tachyarrhythmias. J Cardiovasc
[30] Allessie M, Ausma J, Schotten U. Electrical, contractile and structural remodeling Electrophysiol 1996;7:999–1007.
during atrial fibrillation. Cardiovasc Res 2002;54:230–46. [62] Coumel P, Attuel P, Lavallee J, Flammang D, Leclercq JF, Slama R. The atrial
[31] Falk RH. Atrial fibrillation. N Engl J Med 2001;344:1067–78. arrhythmia syndrome of vagal origin. Arch Mal Coeur Vaiss 1978;71:645–56.
[32] Falk RH. Etiology and complications of atrial fibrillation: insights from pathology [63] Chen YJ, Chen SA, Chang MS, Lin CI. Arrhythmogenic activity of cardiac muscle in
studies. Am J Cardiol 1998;82:10N–7N. pulmonary veins of the dog: implication for the genesis of atrial fibrillation.
[33] Aviles RJ, Martin DO, Apperson-Hansen C, Houghtaling PL, Rautaharju P, Kronmal Cardiovasc Res 2000;48:265–73.
RA, et al. Inflammation as a risk factor for atrial fibrillation. Circulation 2003;108: [64] Singh JP, Larson MG, Levy D, Evans JC, Tsuji H, Benjamin EJ. Is baseline autonomic
3006–10. tone associated with new onset atrial fibrillation?: Insights from the Framingham
[34] Frustaci A, Chimenti C, Bellocci F, Morgante E, Russo MA, Maseri A. Histological heart study. Ann Noninvasive Electrocardiol 2004;9:215–20.
substrate of atrial biopsies in patients with lone atrial fibrillation. Circulation [65] Tomita T, Takei M, Saikawa Y, Hanaoka T, Uchikawa S, Tsutsui H, et al. Role of
1997;96:1180–4. autonomic tone in the initiation and termination of paroxysmal atrial fibrillation in
[35] Nakamura Y, Nakamura K, Fukushima-Kusano K, Ohta K, Matsubara H, Hamuro T, patients without structural heart disease. J Cardiovasc Electrophysiol 2003;14:559–64.
et al. Tissue factor expression in atrial endothelia associated with nonvalvular [66] Nagasawa H, Fujiki A, Fujikura N, Matsuda T, Yamashita T, Inoue H. Effects of a
atrial fibrillation: possible involvement in intracardiac thrombogenesis. Thromb novel class III antiarrhythmic agent, NIP-142, on canine atrial fibrillation and
Res 2003;111:137–42. flutter. Circ J 2002;66:185–91.
[36] Kumagai K, Nakashima H, Saku K. The HMG-CoA reductase inhibitor atorvastatin [67] Coumel P. Atrial fibrillation: one more sporting inconvenience? Eur Heart J
prevents atrial fibrillation by inhibiting inflammation in a canine sterile 2002;23:431–3.
pericarditis model. Cardiovasc Res 2004;62:105–11. [68] Nattel S. Therapeutic implications of atrial fibrillation mechanisms: can mech-
[37] Dernellis J, Panaretou M. C-reactive protein and paroxysmal atrial fibrillation: anistic insights be used to improve AF management? Cardiovasc Res 2002;54:
evidence of the implication of an inflammatory process in paroxysmal atrial 347–60.
fibrillation. Acta Cardiol 2001;56:375–80. [69] Van Gelder IC, Hagens VE, Bosker HA, Kingma JH, Kamp O, Kingma T, et al. A
[38] Chung MK, Martin DO, Sprecher D, Wazni O, Kanderian A, Carnes CA, et al. C-reactive comparison of rate control and rhythm control in patients with recurrent
protein elevation in patients with atrial arrhythmias: inflammatory mechanisms persistent atrial fibrillation. N Engl J Med 2002;347:1834–40.
and persistence of atrial fibrillation. Circulation 2001;104:2886–91. [70] Wyse DG, Waldo AL, DiMarco JP, Domanski MJ, Rosenberg Y, Schron EB, et al. A
[39] Yared JP, Starr NJ, Torres FK, Bashour CA, Bourdakos G, Piedmonte M, et al. Effects comparison of rate control and rhythm control in patients with atrial fibrillation.
of single dose, postinduction dexamethasone on recovery after cardiac surgery. N Engl J Med 2002;347:1825–33.
Ann Thorac Surg 2000;69:1420–4. [71] Hohnloser SH, Kuck KH, Lilienthal J. Rhythm or rate control in atrial fibrillation—
[40] Halonen J, Halonen P, Jarvinen O, Taskinen P, Auvinen T, Tarkka M, et al. Pharmacological Intervention in Atrial Fibrillation (PIAF): a randomised trial.
Corticosteroids for the prevention of atrial fibrillation after cardiac surgery: a Lancet 2000;356:1789–94.
randomized controlled trial. JAMA 2007;297:1562–7. [72] Carlsson J, Miketic S, Windeler J, Cuneo A, Haun S, Micus S, et al. Randomized trial of
[41] Engelmann MD, Svendsen JH. Inflammation in the genesis and perpetuation of rate-control versus rhythm control in persistent atrial fibrillation: the Strategies of
atrial fibrillation. Eur Heart J 2005;26:2083–92. Treatment of Atrial Fibrillation (STAF) study. J Am Coll Cardiol 2003;41:1690–6.
[42] Korantzopoulos P, Kolettis T, Siogas K, Goudevenos J. Atrial fibrillation and [73] Opolski G, Torbicki A, Kosior DA, Szulc M, Wozakowska-Kaplon B, Kolodziej P, et al.
electrical remodeling: the potential role of inflammation and oxidative stress. Rate control vs rhythm control in patients with nonvalvular persistent atrial
Med Sci Monit 2003;9 RA225-9. fibrillation: the results of the Polish How to Treat Chronic Atrial Fibrillation (HOT
[43] Benjamin EJ, Levy D, Vaziri SM, D'Agostino RB, Belanger AJ, Wolf PA. Independent CAFE) Study. Chest 2004;126:476–86.
risk factors for atrial fibrillation in a population-based cohort. The Framingham [74] Fuster V, Ryden LE, Cannom DS, Crijns HJ, Curtis AB, Ellenbogen KA, et al. ACC/
Heart Study. JAMA 1994;271:840–4. AHA/ESC 2006 Guidelines for the Management of Patients with Atrial Fibrilla-
[44] Kato T, Yamashita T, Sekiguchi A, Sagara K, Takamura M, Takata S, et al. What are tion: a report of the American College of Cardiology/American Heart Association
arrhythmogenic substrates in diabetic rat atria? J Cardiovasc Electrophysiol Task Force on Practice Guidelines and the European Society of Cardiology
2006;17:890–4. Committee for Practice Guidelines (Writing Committee to Revise the 2001
[45] Patlolla V, Alsheikh-Ali AA, Al-Ahmad AM. The renin–angiotensin system: a Guidelines for the Management of Patients with Atrial Fibrillation): developed in
therapeutic target in atrial fibrillation. Pacing Clin Electrophysiol 2006;29: 1006–12. collaboration with the European Heart Rhythm Association and the Heart
[46] Kumagai K, Nakashima H, Urata H, Gondo N, Arakawa K, Saku K. Effects of Rhythm Society. Circulation 2006;114:e257–354.
angiotensin II type 1 receptor antagonist on electrical and structural remodeling [75] Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation.
in atrial fibrillation. J Am Coll Cardiol 2003;41:2197–204. Analysis of pooled data from five randomized controlled trials. Arch Intern Med
[47] Pedersen OD, Bagger H, Kober L, Torp-Pedersen C. Trandolapril reduces the 1994;154:1449–1457.
incidence of atrial fibrillation after acute myocardial infarction in patients with [76] Medi C, Hankey GJ, Freedman SB. Atrial fibrillation. Med J Aust 2007;186:197–202.
left ventricular dysfunction. Circulation 1999;100:376–80. [77] Albers GW. Antithrombotic therapy for prevention and treatment of ischemic
[48] Vermes E, Tardif JC, Bourassa MG, Racine N, Levesque S, White M, et al. Enalapril stroke. J Thromb Thrombolysis 2001;12:19–22.
decreases the incidence of atrial fibrillation in patients with left ventricular [78] Verma A, Natale A. Should atrial fibrillation ablation be considered first-line therapy
dysfunction: insight from the Studies Of Left Ventricular Dysfunction (SOLVD) for some patients? Why atrial fibrillation ablation should be considered first-line
trials. Circulation 2003;107:2926–31. therapy for some patients. Circulation 2005;112:1214–22 discussion 1231.
[49] Hobbs WJ, Van Gelder IC, Fitzpatrick AP, Crijns HJ, Garratt CJ. The role of atrial [79] Corley SD, Epstein AE, DiMarco JP, Domanski MJ, Geller N, Greene HL, et al.
electrical remodeling in the progression of focal atrial ectopy to persistent atrial Relationships between sinus rhythm, treatment, and survival in the Atrial
fibrillation. J Cardiovasc Electrophysiol 1999;10:866–70. Fibrillation Follow-Up Investigation of Rhythm Management (AFFIRM) Study.
[50] Wu TJ, Ong JJ, Chang CM, Doshi RN, Yashima M, Huang HL, et al. Pulmonary veins Circulation 2004;109:1509–13.
and ligament of Marshall as sources of rapid activations in a canine model of [80] Falk RH. Management of atrial fibrillation—radical reform or modest modifica-
sustained atrial fibrillation. Circulation 2001;103:1157–63. tion? N Engl J Med 2002;347:1883–4.
[51] Kumagai K, Yasuda T, Tojo H, Noguchi H, Matsumoto N, Nakashima H, et al. Role [81] Chung MK. Atrial fibrillation: rate control is as good as rhythm control for some,
of rapid focal activation in the maintenance of atrial fibrillation originating from but not all. Cleve Clin J Med 2003;70:567–73.
the pulmonary veins. Pacing Clin Electrophysiol 2000;23:1823–7. [82] Pappone C, Rosanio S, Augello G, Gallus G, Vicedomini G, Mazzone P, et al.
[52] Sueda T, Imai K, Ishii O, Orihashi K, Watari M, Okada K. Efficacy of pulmonary vein Mortality, morbidity, and quality of life after circumferential pulmonary vein
isolation for the elimination of chronic atrial fibrillation in cardiac valvular ablation for atrial fibrillation: outcomes from a controlled nonrandomized long-
surgery. Ann Thorac Surg 2001;71:1189–93. term study. J Am Coll Cardiol 2003;42:185–97.
[53] Nathan H, Gloobe H. Myocardial atrio-venous junctions and extensions (sleeves) [83] Crijns HJ. Rate versus rhythm control in patients with atrial fibrillation: what the
over the pulmonary and caval veins. Anatomical observations in various mammals. trials really say. Drugs 2005;65:1651–67.
Thorax 1970;25:317–24. [84] Cooper HA, Bloomfield DA, Bush DE, Katcher MS, Rawlins M, Sacco JD, et al.
[54] Saito T, Waki K, Becker AE. Left atrial myocardial extension onto pulmonary veins Relation between achieved heart rate and outcomes in patients with atrial
in humans: anatomic observations relevant for atrial arrhythmias. J Cardiovasc fibrillation (from the Atrial Fibrillation Follow-up Investigation of Rhythm
Electrophysiol 2000;11:888–94. Management [AFFIRM] Study). Am J Cardiol 2004;93:1247–53.
[55] Perez-Lugones A, McMahon JT, Ratliff NB, Saliba WI, Schweikert RA, Marrouche [85] Bjerregaard P, Bailey WB, Robinson SE. Rate control in patients with chronic atrial
NF, et al. Evidence of specialized conduction cells in human pulmonary veins of fibrillation. Am J Cardiol 2004;93:329–32.
patients with atrial fibrillation. J Cardiovasc Electrophysiol 2003;14:803–9. [86] Kowey PR. You only get so many heartbeats. J Am Coll Cardiol 2004;43:1209–10.
[56] Ho SY, Cabrera JA, Tran VH, Farre J, Anderson RH, Sanchez-Quintana D. Architecture of [87] Van Gelder IC, Rienstra M, Van den Berg MP, Van Veldhuisen DJ. Rate control in
the pulmonary veins: relevance to radiofrequency ablation. Heart 2001;86:265–70. atrial fibrillation. J Am Coll Cardiol 2004;44:2417–8 author reply 2418–9.
S.T. Mathew et al. / European Journal of Internal Medicine 20 (2009) 672–681 681

[88] Wood MA, Brown-Mahoney C, Kay GN, Ellenbogen KA. Clinical outcomes after [108] Morady F. Treatment of paroxysmal atrial fibrillation by pulmonary vein
ablation and pacing therapy for atrial fibrillation: a meta-analysis. Circulation isolation. Circ J 2003;67:567–71.
2000;101:1138–44. [109] Pappone C, Oreto G, Rosanio S, Vicedomini G, Tocchi M, Gugliotta F, et al. Atrial
[89] Dorian P, Connors SP. Pharmacological and nonpharmacological methods for rate electroanatomic remodeling after circumferential radiofrequency pulmonary
control. Can J Cardiol 2005;21(Suppl B):26B–30B. vein ablation: efficacy of an anatomic approach in a large cohort of patients with
[90] Levy T, Walker S, Mason M, Spurrell P, Rex S, Brant S, et al. Importance of rate atrial fibrillation. Circulation 2001;104:2539–44.
control or rate regulation for improving exercise capacity and quality of life in [110] Schwartzman D, Parizhskaya M, Devine WA. Linear ablation using an irrigated
patients with permanent atrial fibrillation and normal left ventricular function: a electrode electrophysiologic and histologic lesion evolution comparison with ablation
randomised controlled study. Heart 2001;85:171–8. utilizing a non-irrigated electrode. J Interv Card Electrophysiol 2001;5:17–26.
[91] Weerasooriya R, Davis M, Powell A, Szili-Torok T, Shah C, Whalley D, et al. The [111] Ho SY, Sanchez-Quintana D, Cabrera JA, Anderson RH. Anatomy of the left
Australian Intervention Randomized Control of Rate in Atrial Fibrillation Trial atrium: implications for radiofrequency ablation of atrial fibrillation. J Cardiovasc
(AIRCRAFT). J Am Coll Cardiol 2003;41:1697–702. Electrophysiol 1999;10:1525–33.
[92] Ahmad K, Dorian P. Rate control in atrial fibrillation: looking beyond the average [112] Seidl K, Schwacke H, Zahn R, Rameken M, Drogemuller A, Senges J. Catheter ablation
heart rate. Curr Opin Cardiol 2006;21:88–93. of chronic atrial fibrillation with noncontact mapping: are continuous linear lesions
[93] Bailin SJ, Adler S, Giudici M. Prevention of chronic atrial fibrillation by pacing in associated with ablation success? Pacing Clin Electrophysiol 2003;26:534–43.
the region of Bachmann's bundle: results of a multicenter randomized trial. [113] Ernst S, Ouyang F, Goya M, Lober F, Schneider C, Hoffmann-Riem M, et al. Total
J Cardiovasc Electrophysiol 2001;12:912–7. pulmonary vein occlusion as a consequence of catheter ablation for atrial fib-
[94] Padeletti L, Porciani MC, Michelucci A, Colella A, Costoli A, Ciapetti C, et al. rillation mimicking primary lung disease. J Cardiovasc Electrophysiol 2003;14:
Prevention of short term reversible chronic atrial fibrillation by permanent 366–70.
pacing at the triangle of Koch. J Interv Card Electrophysiol 2000;4:575–83. [114] Keane D, Mansour M, Singh J. Detection by intracardiac echocardiography of
[95] Hermida JS, Kubala M, Lescure FX, Delonca J, Clerc J, Otmani A, et al. Atrial septal early formation of left atrial thrombus during pulmonary vein isolation. Europace
pacing to prevent atrial fibrillation in patients with sinus node dysfunction: 2004;6:109–10.
results of a randomized controlled study. Am Heart J 2004;148:312–7. [115] Schwab JO, Burkhardt D, Yang A, Schrickel J, Luderitz B, Lewalter T. ECG signs
[96] Doshi RN, Daoud EG, Fellows C, Turk K, Duran A, Hamdan MH, et al. Left mimicking acute inferior wall myocardial infarction are associated with elevated
ventricular-based cardiac stimulation post AV nodal ablation evaluation (the myocardial enzymes during isolation of pulmonary vein for focal atrial fibrillation.
PAVE study). J Cardiovasc Electrophysiol 2005;16:1160–5. Europace 2004;6:111–5.
[97] Cox JL, Boineau JP, Schuessler RB, Jaquiss RD, Lappas DG. Modification of the maze [116] Packer DL, Asirvatham S, Munger TM. Progress in nonpharmacologic therapy of
procedure for atrial flutter and atrial fibrillation. I. Rationale and surgical results. atrial fibrillation. J Cardiovasc Electrophysiol 2003;14:S296–309.
J Thorac Cardiovasc Surg 1995;110:473–84. [117] Patterson E, Po SS, Scherlag BJ, Lazzara R. Triggered firing in pulmonary veins
[98] Gillinov AM, McCarthy PM. Advances in the surgical treatment of atrial initiated by in vitro autonomic nerve stimulation. Heart Rhythm 2005;2:624–31.
fibrillation. Cardiol Clin 2004;22:147–57. [118] Marron K, Wharton J, Sheppard MN, Fagan D, Royston D, Kuhn DM, et al.
[99] Damiano Jr RJ, Gaynor SL, Bailey M, Prasad S, Cox JL, Boineau JP, et al. The long- Distribution, morphology, and neurochemistry of endocardial and epicardial
term outcome of patients with coronary disease and atrial fibrillation undergoing nerve terminal arborizations in the human heart. Circulation 1995;92:2343–51.
the Cox maze procedure. J Thorac Cardiovasc Surg 2003;126:2016–21. [119] Kaye DM, Vaddadi G, Gruskin SL, Du XJ, Esler MD. Reduced myocardial nerve
[100] Chen SA, Hsieh MH, Tai CT, Tsai CF, Prakash VS, Yu WC, et al. Initiation of atrial growth factor expression in human and experimental heart failure. Circ Res
fibrillation by ectopic beats originating from the pulmonary veins: electro- 2000;86:E80–4.
physiological characteristics, pharmacological responses, and effects of radio- [120] Ginty DD, Segal RA. Retrograde neurotrophin signaling: Trk-ing along the axon.
frequency ablation. Circulation 1999;100:1879–86. Curr Opin Neurobiol 2002;12:268–74.
[101] Haissaguerre M, Shah DC, Jais P, Hocini M, Yamane T, Deisenhofer I, et al. [121] Cao JM, Fishbein MC, Han JB, Lai WW, Lai AC, Wu TJ, et al. Relationship between
Electrophysiological breakthroughs from the left atrium to the pulmonary veins. regional cardiac hyperinnervation and ventricular arrhythmia. Circulation
Circulation 2000;102:2463–5. 2000;101:1960–9.
[102] Haissaguerre M, Jais P, Shah DC, Garrigue S, Takahashi A, Lavergne T, et al. [122] Kim DT, Luthringer DJ, Lai AC, Suh G, Czer L, Chen LS, et al. Sympathetic nerve sprouting
Electrophysiological end point for catheter ablation of atrial fibrillation initiated after orthotopic heart transplantation. J Heart Lung Transplant 2004;23:1349–58.
from multiple pulmonary venous foci. Circulation 2000;101:1409–17. [123] Cao JM, Chen LS, KenKnight BH, Ohara T, Lee MH, Tsai J, et al. Nerve sprouting and
[103] Pappone C, Rosanio S, Oreto G, Tocchi M, Gugliotta F, Vicedomini G, et al. sudden cardiac death. Circ Res 2000;86:816–21.
Circumferential radiofrequency ablation of pulmonary vein ostia: a new [124] Swissa M, Zhou S, Gonzalez-Gomez I, Chang CM, Lai AC, Cates AW, et al. Long-
anatomic approach for curing atrial fibrillation. Circulation 2000;102:2619–28. term subthreshold electrical stimulation of the left stellate ganglion and a canine
[104] Gerstenfeld EP, Callans D, Dixit S, Lin D, Cooper J, Russo AM, et al. Characteristics model of sudden cardiac death. J Am Coll Cardiol 2004;43:858–64.
of patients undergoing atrial fibrillation ablation: trends over a seven-year [125] Lafuente-Lafuente C, Mouly S, Longas-Tejero MA, Mahe I, Bergmann JF.
period 1999–2005. J Cardiovasc Electrophysiol 2007;18:23–8. Antiarrhythmic drugs for maintaining sinus rhythm after cardioversion of atrial
[105] Haissaguerre M, Shah DC, Jais P, Hocini M, Yamane T, Deisenhofer I, et al. fibrillation: a systematic review of randomized controlled trials. Arch Intern Med
Mapping-guided ablation of pulmonary veins to cure atrial fibrillation. Am J 2006;166:719–28.
Cardiol 2000;86:9K–19K. [126] Page RL. Medical management of atrial fibrillation: future directions. Heart
[106] Keane D, Ruskin J. Pulmonary vein isolation for atrial fibrillation. Rev Cardiovasc Rhythm 2007;4:S91–4.
Med 2002;3:167–75. [127] Olsson SB. Atrial fibrillation—new aspects on mechanism and treatment. J Intern
[107] Oral H, Knight BP, Tada H, Ozaydin M, Chugh A, Hassan S, et al. Pulmonary vein Med 1996;239:3–15.
isolation for paroxysmal and persistent atrial fibrillation. Circulation 2002;105:
1077–81.

You might also like