Next Article in Journal
Predicting Outcome in Patients with Brain Injury: Differences between Machine Learning versus Conventional Statistics
Previous Article in Journal
In Vitro and Ex Vivo Evaluation of Mangifera indica L. Extract-Loaded Green Nanoparticles in Topical Emulsion against Oxidative Stress and Aging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Percutaneous Treatment Approaches in Atrial Fibrillation: Current Landscape and Future Perspectives

by
Panagiotis Theofilis
1,
Evangelos Oikonomou
2,
Alexios S. Antonopoulos
1,
Gerasimos Siasos
2,
Konstantinos Tsioufis
1 and
Dimitris Tousoulis
1,*
1
First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
2
Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(9), 2268; https://doi.org/10.3390/biomedicines10092268
Submission received: 11 August 2022 / Revised: 3 September 2022 / Accepted: 9 September 2022 / Published: 13 September 2022

Abstract

:
Atrial fibrillation (AF), the most common sustained arrhythmia in clinical practice, represents a major cause of morbidity and mortality, with an increasing prevalence. Pharmacologic treatment remains the cornerstone of its management through rhythm and rate control, as well as the prevention of thromboembolism with the use of oral anticoagulants. Recent progress in percutaneous interventional approaches have provided additional options in the therapeutic arsenal, however. The use of the different catheter ablation techniques can now lead to long arrhythmia-free intervals and significantly lower AF burden, thus reducing the rate of its complications. Particularly encouraging evidence is now available for patients with persistent AF or concomitant heart failure, situations in which catheter ablation could even be a first-line option. In the field of stroke prevention, targeting the left atrial appendage with percutaneous device implantation may reduce the risk of thromboembolism to lower rates than that predicted with conventional ischemic risk scores. Left atrial appendage occlusion through the approved Watchman or Amplatzer devices is a well-established, efficacious, and safe method, especially in high-ischemic and bleeding risk patients with contraindications for oral anticoagulation.

1. Introduction

The prevalence and incidence rates of atrial fibrillation (AF), the most common sustained arrhythmia in clinical practice, are constantly rising, especially in developed, high-income countries [1]. Over 400 new cases per million persons were reported in 2017, displaying a significant increase compared to the 345 and 309 new cases per million persons that were observed in 2007 and 1997, respectively [1]. As a result, the worldwide AF prevalence is approximately 37.574 million cases, representing a 33% increase compared to 1997 [1]. Additionally, there has been a 105% increase in deaths attributable to AF from 1997 to 2017 (0.51% of overall mortality) [1]. These trends will continue to predominate in the upcoming decades, with an absolute increase in incidence and prevalence of approximately 63% and 66%, respectively [1]. Major modifiable and non-modifiable risk factors should be taken into account for this increase, such as population ageing, female sex, arterial hypertension, obesity, diabetes mellitus, and genetics, among others [2,3,4,5].
AF is responsible for important life-threatening complications, such as embolic stroke and heart failure (HF) [6,7], and studies have also shown a decrease in cognitive function in subjects with AF [8]. Ultimately, individuals with AF may have depression and impaired quality of life [9,10], as well as a higher rate of hospitalizations and mortality [11]. However, the institution of novel therapeutic approaches is believed to ameliorate patient prognosis. In particular, interventional approaches are gaining ground in the management algorithms, owing to the encouraging data from clinical trials. This review aims to summarize the current knowledge and applications of contemporary percutaneous procedures in the management of AF, namely catheter ablation (CA) and left atrial appendage closure (LAAC).

2. Atrial Fibrillation Catheter Ablation

Early rhythm control is essential in reducing the burden of AF complications [12], regardless of the presence of symptoms [13]. Percutaneous catheter AF ablation constitutes an attractive approach towards rhythm control. Radiofrequency (RF) CA, in particular, is the most commonly performed ablation procedure in electrophysiology. It predominantly consists of pulmonary vein isolation (PVI), as these are considered major initiators of paroxysmal AF [14]. After this first description in 1998, segmental ostial PV ablation was introduced and was bolstered by the use of 3D electroanatomical mapping [15]. Later on, wide-area circumferential ablation with verification of the conduction block was proven superior than ostial segmental isolation of each individual pulmonary vein [16,17], through the neutralization of more proximal trigger sources and providing concomitant autonomic denervation [18]. Consequently, antral ablation is the main AF ablation technique and is recommended during all AF ablation procedures [19]. In CA procedures for AF, uninterrupted periprocedural anticoagulation with direct oral anticoagulants is suggested [20]. Moreover, the use of intracardiac echocardiography could also be of use for reducing fluoroscopy time, procedure duration, and the rate of complications [21].
Regarding clinical outcomes, CA reduces the risk of stroke/transient ischemic attack (risk ratio (RR) 0.61, 95% CI 0.39–0.97, p = 0.035) and death (RR 0.7, 95% CI 0.55–0.89, p = 0.004), compared to medical treatment [22]. Moreover, it may lead to a greater rate of sinus rhythm maintenance (RR 3.55, 95% CI 2.34–5.40, p < 0.001) and improve the left ventricular ejection fraction (weighted mean difference (WMD) 5.39, 95% CI 2.45–8.32, p < 0.001) [22]. Critically, CA for AF appears to be a cost-effective procedure, according to an analysis performed in the United Kingdom [23]. It should be noted that the large-scale CABANA randomized clinical trial, comparing CA to anti-arrhythmic drugs (AADs) in AF, was neutral regarding the primary composite end point of death, disabling stroke, serious bleeding, or cardiac arrest. Concerns about the lower-than-expected event rates and treatment crossovers have been raised, and, therefore, the results should be cautiously considered [24]. Moreover, age variations were detected, with younger age populations (<65 years of age) exhibiting the greatest relative and absolute benefits from the CA procedure [25].
In the landmark trial mentioned above, AF recurrence was still significantly reduced by CA compared to AADs during the 5-year follow-up, irrespective of AF type [26]. However, AF recurrence after CA is not uncommon. Several factors are believed to be implicated, including structural heart disease, left atrium characteristics (diameter, voltage), PV isolation-reconnections, AF duration, the presence of non-PV triggers, cardiometabolic risk factors, chronic kidney disease, and anxiety [27,28,29]. Interestingly, AF ablation in the CABANA randomized clinical trial led to reductions in left atrial volume index and mean PV ostial diameter, which were associated with lower AF recurrence rates [30]. Biomarkers have also been associated with AF recurrence after CA, namely N-Terminal-pro-B-type-natriuretic peptide, B-type natriuretic peptide, high-sensitivity C-reactive protein, carboxy-terminal telopeptide of collagen type I, interleukin-6, and galectin-3 [31,32]. However, their exact role in patients undergoing CA for AF needs to be precisely determined.
As endorsed by the latest guidelines from the European Society of Cardiology and the European Heart Rhythm Association, published in 2020, CA is now a mainstay of AF rhythm control strategies [33]. It is recommended as a first line treatment option in patients with suspected AF-induced cardiomyopathy. It should be considered in individuals with symptomatic paroxysmal AF or with concomitant AF and HF with a reduced left ventricular ejection fraction. Lastly, it can be considered in patients with persistent AF in the absence of risk factors for recurrence. Moreover, in cases of drug failure or intolerance, CA is recommended irrespective of the AF type.

2.1. Catheter Ablation Types

2.1.1. Radiofrequency Ablation

The effectiveness of RF ablation has been established. This procedure was associated with greater arrhythmia-free survival (RR 0.62, 95% CI 0.51–0.74, p < 0.001) and less hospitalizations (RR 0.32, 95% CI 0.19–0.53, p < 0.001) compared to AADs in a recent meta-analysis [34]. Other than the effectiveness in reducing major endpoints, a reduced risk of incident dementia with AF CA was proposed by Saglietto et al. in their systematic review and meta-analysis [35]. Moreover, it may also prevent the progression of paroxysmal to persistent AF [36]. This is particularly important, since such patients may be facing an adverse cardiovascular prognosis, as previously shown [37]. It should be noted that same-day discharge may be feasible, safe, and effective in selected patients [38]. Ablation guidance through unipolar signal modification may improve PVI outcomes in terms of lower radiofrequency time and AF recurrence at 12 months, as shown in a randomized clinical trial of 136 patients with paroxysmal AF [39].
The electrical reconnection of at least one PV may influence the arrhythmia-free interval after an RF ablation procedure. Interestingly, it should be noted that the presence of electrical reconnections in those patients undergoing a redo RF ablation may be a good predictive sign compared to those without PV reconnections, as the number of reconnected PVs was independently related to lower AF recurrence in a mean 18.4-month follow-up (hazard ratio (HR) 0.56, 95% confidence interval (CI) 0.34–0.95, p = 0.032) [40]. To manage PV reconnection, operators could opt for a contact force of 5–10 g, and assess for the loss of capture over the ablation line, confirm the exit block, and perform adenosine testing [19]. Improvement in mapping techniques may further support the successful PVI with the use of high-density multielectrode catheters, providing more precise electroanatomical maps that reflect real-time volume-rendered left atrium and PV geometry during ablation. In a recent study, the high-density grid catheter, consisting of four splines, each with four small (1 mm), equally spaced (3 mm) electrodes, was able to detect a significantly higher number of PV reconnections compared to the standard bipolar mapping [41]. For the lesion size, minimum ablation indexes (AI) of ≥370 and ≥480 for posterior-inferior and anterior-superior segments, respectively, should be sought [42]. Other protocols, such as the CLOSE protocol which incorporates AI, inter-lesion distance, and catheter stability, have been proven reliable in achieving adequate PVI with an impressive rate of freedom from atria tachyarrhythmia [43]. Moreover, lesion size index is another metric attributed to the TactiCath ablation catheter, with higher values being associated with a shorter procedure, radiofrequency, and fluoroscopy times, as well as fewer touch-up ablations compared to the low lesion size index [44].
Regarding the additional ablation of AF triggers (Figure 1), the performance of circumferential PVI together with an electrical posterior box isolation in subjects with reconnected PV did not confer any additional efficacy regarding arrhythmia-free survival [45]. Posterior left atrial wall isolation is under investigation in other ongoing clinical trials of AF ablation [46,47], with the results being expected to provide essential information. Regarding other ablation sites, targeting ectopy-triggering ganglionated plexuses was equally effective to PVI in patients with paroxysmal AF, with a trend towards a lower rate of atrial arrhythmia prevention and significantly lower AAD usage after the procedure [48]. This finding was further supported by a meta-analysis, which showed a lower arrhythmia recurrence (odds ratio (OR) 0.58, 95% CI 0.41–0.82) that was dependent on the left atrial diameter in a meta-regression analysis [49]. Lately, Li et al. showed that the vein of Marshall ethanol infusion decreased atrial tachyarrhythmia recurrence compared to PVI alone, especially when substrate ablation was also performed [50].
A modified high-power, short-duration (HPSD) RF ablation protocol has also been proposed, aiming to improve lesion-to-lesion uniformity and produce a transmural injury [51]. In the earliest study of Nilsson et al., the use of power output at 45 W for 20 s resulted in a greater need for more RF applications to achieve adequate PVI, with a shorter procedural time compared to a 30 W/120 s duration conventional CA [52]. In a subsequent randomized trial, HPSD was associated with a lower number of ablations needed for PVI, however [53]. HPSD ablation appears to be a more durable procedure, with lower rates of chronic PV reconnection, owing to enhanced catheter stability [54]. This finding was not accompanied with lower rates of arrhythmia-free intervals, however. This could be associated with progressive fibrous atrial cardiomyopathy, which may be evident in the long term after HPSD [55]. Posterior box and superior vena cava isolation are also feasible with a HPSD ablation method [56,57]. Additionally, HPSD is an appropriate option in redo CA procedures, as shown by Junarta et al. [58]. According to meta-analytic evidence, HPSD ablation results in a greater rate of freedom from atrial arrhythmia (OR 1.44), decreased acute PV reconnection (OR 0.56), and ameliorated first-pass PVI (OR 3.58) [59]. Moreover, the procedural, fluoroscopy, and ablation times were all significantly reduced, paired with similar rates of complications compared to conventional RF ablation [59]. Concerning the procedural safety, the operators should avoid the delivery of lesions in quick succession, as this may induce esophageal overheating and injury. Esophageal thermal injury is more common in subjects undergoing HPSD RF ablation procedures, but are not related to esophageal lesions in esophagogastroduodenoscopy [60,61,62]. Even in subjects exceeding luminaλ esophageal temperatures of 39 °C, there were no esophageal perforations or atrial–esophageal fistulas after upper endoscopic evaluation [63]. AI guidance or the CLOSE protocol could be safely used in HPSD ablation procedures, as previously shown [64].
The very high-power, short-duration (vHPSD) RF ablation technique involves a power output of 90 W for 4 s, with acceptable safety and efficacy proven in the QDOT-FAST trial [65], subsequently confirmed in the fast and furious AF study [66]. In a study with slightly lower thresholds (70 W/7 s), vHPSD led to a greater rate of arrhythmia-free intervals at 1 year, as well as decreased RF and procedural time, compared to the conventional procedure [67]. When incorporating temperature control during vHPSD ablation, there was no evidence of postprocedural esophageal ulceration, with silent cerebral events being noted in 24% of patients [68].

2.1.2. Cryoablation

Other than RF ablation, cryoablation through the use of a cryoballoon is a valid alternative approach. This method aims to create intramural, irreversible myocardial lesions, and is characterized by a short learning curve [69]. Regarding this technique, the single application of the cryoballoon at −40 °C and the achievement of isolation within 60 s are predictors of adequate electrical PVI [70]. Moving to clinical outcomes, in a recently reported randomized clinical trial of patients with symptomatic, paroxysmal AF, cryoablation performance was associated with a significantly lower atrial tachyarrhythmia recurrence (HR 0.48, 95% CI 0.35–0.66, p < 0.001) [71], confirming other lines of evidence [72,73]. Moreover, this technique may also lead to significant improvement in the quality of life, as well as to symptom resolution [74,75]. However, early arrhythmia recurrence in the 3-month blanking period was associated with a lower arrhythmia-free survival in patients undergoing cryoballoon ablation [76]. Comparative studies between RF and cryoablation have been performed to assess the most effective and safe procedure. In a recently reported randomized clinical trial of 346 patients with paroxysmal AF, there was no difference in the incidence of symptomatic atrial arrhythmia with the use of contact force–guided RF ablation, 4-min cryoablation, or 2-min cryoablation [77]. Moreover, all of those regimens appeared equally safe, with RF ablation being a longer procedure with shorter fluoroscopy time [77]. Interestingly, cryoballoon ablation also had a shorter procedural duration compared to HPSD RF ablation [78]. A recent analysis also pointed to potential sex-related differences in the outcomes between cryoballoon and RF ablation, with men exhibiting a better efficacy with cryoablation, with procedure failure defined as the recurrence of atrial arrhythmia, reablation, and reinitiation of anti-arrhythmic medication [79].

2.1.3. Laser Balloon Ablation

Laser balloon (LB), through the use of point-by-point laser energy and covering 30° of a circle with each lesion, is another alternative with equal efficacy and safety to RF and cryoballoon ablation techniques [80,81,82]. Interestingly, visually-guided LB ablation was associated with lower atrial arrhythmia recurrence at 12 months compared to RF ablation in one study, with a negative adenosine provocation test being predictive of a positive outcome [83]. This could be related to a more complete PVI isolation and a lesser extent of fibrosis, as demonstrated by a previous cardiac magnetic resonance imaging study [84]. Additionally, a wide-area circumferential LB ablation led to a greater arrhythmia-free interval compared to a wide-area circumferential RF ablation in a retrospective multicenter study [85]. Moreover, a manual dragging laser irradiation technique could shorten the procedure duration without compromising efficacy or safety, as shown in the study of Sasaki et al. [86]. Third-generation LB ablation is characterized by a shorter laser application and procedure duration compared to second-generation, with a similar effect [87]. In another study of patients requiring redo procedures with second- and third-generation LB due to AF recurrence, persistent PVI was more frequent in the third-generation LB [88].

2.1.4. Pulsed Field Ablation

The application of electroporation in the context of pulsed field ablation (PFA) is an evolving method in the field of PVI through CA. By inducing irreversible electroporation, durable lesions may be formed due to the loss of cellular homeostasis. Moreover, since cardiomyocytes have low sensitivity to PFA, a minimal risk of collateral damage is present. As shown in a recent imaging study, PFA results in an increased late gadolinium enhancement without injuring the microvasculature or inducing intramural hemorrhage, whereas the chronic fibrotic changes are of a lesser magnitude compared to thermal ablation techniques [89]. Additionally, the lack of coagulative necrosis with PFA suggests a minimal risk of incident pulmonary vein stenosis, as demonstrated by Kuroki et al. [90]. Furthermore, a short procedural duration should be mentioned. PFA is also effective in the case of irregular surfaces, where electrode–tissue contact is not ideal, providing more uniform lesions compared to RF ablation [91].
PFA has been evaluated preclinically, with a good efficacy and safety profile [92]. Therefore, clinical data are now accumulating regarding its use in humans. Among the first published reports, Reddy et al. utilized the endocardial catheter, Farawave (Farapulse Inc., Menlo Park, CA, USA). Being the only validated PFA system, this 12F catheter has a distal portion consisting of five splines, each with four electrodes per spline, able to provide voltages between 900 and 2500 V. The PFA and fluoroscopy duration was 19 and 12 min on average, respectively. The PVI was successful in all 15 patients included in this study [93]. According to the IMPULSE and PEFCAT trials that followed, there was a durable PVI in 100% of the procedures at the 3-month follow-up, whereas the freedom from AF was estimated at 87.4% at 1 year [94]. Moreover, the rate of short- and mid-term adverse events was very low. In the latest report of the MANIFEST-PF registry, PVI with PFA was accomplished in 99.9% of patients, with a low rate of major complications [95]. However, antral PVI may not be adequately achieved through PFA, since an insufficient isolation of the left anterior antral PV segments and enlarged LA isolation areas on the posterior segments was detected by Bohnen et al. [96]. Head-to-head trials comparing the different AF CA techniques are essential to demonstrate differences in the efficacy and safety, and to decide on the optimal ablation method.
Other PFA catheters are also available at earlier stages of development, however. To begin with, the circular PVI catheter by the name of PulseSelect (Medtronic, Minneapolis, MN, USA) possesses nine electrodes for bipolar, biphasic energy delivery and electrogram recording, while also being connected to a custom-built PFA or to an RF generator. In the recently published PULSED-AF pilot trial, electrical isolation was achieved in all 152 PV, with a mean procedure duration of 160 min, and without adverse events [97]. Moving to the lattice-tip ablation catheter with a compressible 9-mm nitinol tip, its ability to provide either PFA or RF ablation depending on the morphology of the desired lesion has been demonstrated in a recent pilot study [98]. Finally, a novel multipolar PFA catheter with the ability of real-time PV signal recording appears promising, with similar accuracy to the standard Farawave 3D-mapping system in detecting PVI and residual PV conduction [99]. However, large-scale studies are needed to obtain high-quality evidence for the importance of those systems.

2.1.5. Hot Balloon Ablation

CA of AF through the use of an RF hot balloon is another alternative approach to RF ablation, with early studies showing appropriate PVI, as well as posterior left atrium isolation. A 92% freedom from arrhythmia without the need for AADs, after a mean 11-month follow-up period, was reported [100]. In the long-term, this technique is associated with freedom from atrial tachyarrhythmias in 64.7% of the cases after a 6.2-year follow-up [101]. Re-ablation was mostly required for the PV and posterior left atrium, with the rate of freedom from atrial tachyarrhythmias being 84.5% during the 4.6-year follow-up. PV stenosis and phrenic nerve palsy were infrequent. A single-shot energy application protocol for PV antrum ablation, sparing the PV ostium, has also been tried, with a high rate of sinus rhythm maintenance at 1 year [102]. Some additional hints may enhance the efficacy and safety of the procedure. Real-time balloon surface temperature monitoring when performing single-shot PVI is useful, as temperatures above 58.7 °C may be indicative of appropriate PVI [103]. Regarding safety, esophageal cooling with an infusion of lopamidol and saline in cases of luminal esophageal temperature exceeding 39 °C should be considered to reduce the risk of esophageal injury [104]. Lastly, we should note that although the efficacy of the hot balloon and cryoballoon in terms of AF recurrence is similar, a greater need for touch-up ablation was noted with the hot balloon, especially in left superior pulmonary veins [105]. This was accompanied by a significantly longer procedure duration. A balloon temperature of 73 °C in left superior pulmonary vein ablation may improve the outcome and eliminate the need for touch-up ablation in this region [106].

2.2. Catheter Ablation in Persistent Atrial Fibrillation

Regarding patients with persistent AF, long-term follow-up (~54 months) revealed that RF ablation may reduce the risk of ischemic cerebrovascular events and congestive HF, together with improvements in quality of life, when compared to pharmacotherapy [107]. LB ablation is another alternative in patients with persistent AF, showing similar rates of atrial tachyarrhythmia recurrence to cryoablation in a recently reported propensity-matched analysis [82]. Additional ablation of mapped low voltage areas on top of circumferential PVI did not seem to provide an incremental benefit at 18 months in a randomized trial of patients with persistent AF, since no difference in arrhythmia-free interval was documented [108]. However, in another study, low-voltage area substrate modification in patients with persistent AF undergoing CA resulted in greater arrhythmia-free survival, without any difference in procedure duration or periprocedural complications [109]. Complex-fractionated atrial electrograms (CFAEs) are electrograms with highly fractionated potentials or with a very short cycle length (<120 ms), which represent potential AF substrate sites requiring ablation. The combination of CFAE ablation with high-density voltage mapping resulted in higher rates of freedom from AF compared to PVI alone [110]. Further documentation of CFAE ablation efficacy was present in another study, where the combination of PVI, CFAE, and linear ablation resulted in a greater freedom from atrial arrhythmia in comparison to sole PVI (HR 1.56, 95% CI 1.04–2.34) [111]. Other studies failed to show a difference in 1-year freedom from atrial tachyarrhythmia with CFAE ablation, however [112,113]. This could be attributed to the focal energy sources being located mostly at the border or even outside of the CFAE areas, with those unablated sites potentially leading to AF recurrence [114]. Active CFAEs should be, therefore, distinguished from bystanding CFAEs, potentially through the use of nonlinear recurrence quantification analysis [115]. Active CFAEs characteristically present with an increase in electrogram conformation recurrence [115]. Future adequately designed studies might improve our knowledge in this regard.
Moving to the ablation of posterior atrial wall box isolation on top of circumferential PVI, no effect on the clinical outcomes was noted [116]. However, a recent meta-analysis showed that posterior atrial wall isolation could reduce AF recurrence compared to the control group in patients with persistent AF [117]. Valderrábano et al. investigated the retrograde ethanol infusion in the vein of Marshall, which is perceived as an AF triggering point, and showed that patients with persistent AF randomized to this procedure had a lower rate of atrial tachyarrhythmia recurrence and AF burden compared to conventional RF ablation, without any excess adverse events [118]. Interestingly, thoracoscopic ablation was not proven superior in the group of patients with persistent AF compared to CA regarding freedom from atrial tachyarrhythmia, with the latter being more cost-effective, improving patient symptoms, and providing more quality-adjusted life-years [119]. Lately, magnetic resonance imaging-guided atrial fibrosis ablation, together with PVI, was attempted in patients with persistent AF [120]. The study, however, did not achieve a greater arrhythmia-free interval, and the evaluated procedure was accompanied with more adverse events [120].
Focal, organized rotational activity believed to sustain AF and targeted ablation procedures have, thus, been attempted. This technique may not be appropriate as a sole strategy for paroxysmal AF, since a focal impulse and rotor mapping (FIRM)-guided rotor ablation resulted in shorter arrhythmia-free interval compared to cryoballoon or RF PVI in patients with paroxysmal AF [121,122]. In a study of 58 patients with nonparoxysmal AF, a single FIRM-guided rotor ablation, all patients had identifiable stable atrial rotors [123]. Their ablation resulted in atrial tachyarrhythmia freedom in 73.1% of the patients at 1-year follow-up [123]. FIRM-guided ablation may also be superior to linear left atrial ablation and CFAE ablation regarding the recurrence of AF, as shown by Hsieh et al. [124]. However, a meta-analysis of head-to-head studies comparing PVI with and without FIRM-guided ablation showed no statistically significant difference in atrial tachyarrhythmia recurrence at a mean 18.8 months of follow-up [125]. As the importance of this method is not well established, further evidence from randomized clinical trials may aid us in determining the role of this procedure in persistent AF ablation, possibly through identifying subgroups that might specifically benefit from it.
Dominant frequency (DF) assessment could potentially be of importance in identifying AF-driving regions. However, in the RADAR-AF trial, high-DF area ablation did not have an additive efficacy compared to PVI alone in patients with persistent AF [126]. A recent study found that high-DF areas frequently overlap with low-voltage areas, which are associated with AF recurrence after PVI in patients with nonparoxysmal AF [127]. Ablation of these overlapping areas in conjunction with PVI may lead to greater arrhythmia freedom rates, whereas this decreased in the PVI-only group proportionally to the extent of the low-voltage areas [128]. Since DF is spatiotemporally unstable, the study of Li et al. highlighted that high-DF pattern recurrence is associated with a greater degree of organization [129]. Thus, ablating these recurring patterns may improve the outcomes in patients with persistent AF. Since the optimal use of high-DF patterns in the ablation strategies has not been determined, further studies are required.
In a recently reported Bayesian network meta-analysis of 3394 persistent AF patients and 22 ablation strategies, a strategy involving PVI with left atrial posterior wall and non-PV trigger ablation was characterized as the most effective regarding freedom from atrial tachyarrhythmia recurrence [130]. Concomitant hybrid surgical and CA could also emerge as the most effective alternative in patients with nonparoxysmal AF, despite the increased rate of major adverse events and procedure duration [131]. These observations were mostly based on the landmark CONVERGE randomized controlled trial, which evaluated 149 patients with persistent AF who underwent either a minimally invasive epicardial/endocardial ablation approach or CA. After 1 year of follow-up, the hybrid convergent technique led to significantly lower rates of atrial tachyarrhythmia recurrence, whereas 74% had a ≥90% AF burden reduction [132]. It is clear, however, that the achievement of a favorable outcome in the group of patients with persistent AF is challenging, and further adequately designed studies are required to improve our understanding regarding the optimal ablation strategy.
Finally, we should mention that the evolution of cardiac mapping with novel high-density mapping systems using multielectrode catheters could further improve the outcomes of AF CA in the future, especially in the setting of nonparoxysmal AF. Available systems, such as the Rhythmia HDx ultra-HD, the EnSite Precision, the CARTO 3, and the noncontact AcQMap High Resolution Imaging and Mapping System, may improve AF substrate delineation, greatly improve the efficiency of the mapping procedure without affecting safety, and provide additional AF mechanistic insights [133].

2.3. Catheter Ablation in Heart Failure

CA of AF in HF patients has gained popularity after the presentation of the landmark CASTLE-AF trial (Table 1). Patients with symptomatic AF, HF and a left ventricular ejection fraction of <35%, New York Heart Association class II-IV, and an implantable defibrillator were enrolled. A previous failed response to AADs, the presence of important side effects, or an unwillingness to take these agents were other inclusion criteria. Patients who underwent the CA procedure had a lower risk of facing the composite endpoint of all-cause mortality or HF hospitalization (HR 0.62, 95% CI 0.43–0.87, p = 0.007), after a median 37.8-month follow-up [134]. The effect was irrespective of the degree of left ventricular systolic impairment, and the performance of AF ablation resulted in greater odds of the improvement of left ventricular ejection fraction to over 35% [135]. Additionally, subjects with the less severe functional status exhibited the greatest improvement [135]. Another important aspect of this trial was the AF burden. Though similar at baseline and non-predictive of incident endpoints, the 6-month AF burden was significantly reduced in patients undergoing CA [136]. An AF burden of <50% was associated with a lower incidence of the primary endpoint (HR 0.33, 95% CI 0.15–0.71, p = 0.014) [136]. However, it appears that only a limited number of real-world HF patients with AF meet the trial inclusion criteria [137]. Despite that fact, patients not meeting the inclusion criteria may have a modest benefit from CA (HR 0.79, 95% CI 0.73–0.86, p < 0.001) [137]. On the other hand, patients meeting the exclusion criteria gain no benefit from CA (HR 0.97, 95% CI 0.81–1.17) [137].
CA was also evaluated in HF patients with a high burden of AF, characterized as either paroxysmal AF with more than four episodes in the previous 6 months, or persistent AF with a duration of less than 3 years. The performance of AF ablation was accompanied by an improvement in left ventricular systolic function and quality of life, as well as with a decrease in N-terminal pro-brain natriuretic peptide compared to rate control [138]. A trend towards a lower incidence of the primary endpoint was also noted with AF ablation (HR 0.71, 95% CI 0.49–1.03, p = 0.066) [138]. In another randomized clinical trial, CAMERA-MRI, CA in patients with persistent AF and systolic dysfunction led to ameliorated or even normalized left ventricular ejection fraction, especially in subjects without evidence of ventricular fibrosis [140]. The rate of AF recurrence and burden was 57% and 10.6%, respectively [140]. In the randomized AMICA trial, the improvements in left ventricular systolic function, natriuretic peptides, and quality of life were similar between CA and best medical therapy, despite a numerically lower AF burden [141]. In the less well-studied group of patients with HF and a preserved ejection fraction, CA may be equally effective at maintaining sinus rhythm compared to patients without HF, with a tendency to greater sinus rhythm maintenance and reduced HF hospitalizations compared to medical therapy [142,143].

3. Left Atrial Appendage Closure

Oral anticoagulation remains the cornerstone of stroke prevention in AF patients, with direct oral anticoagulants (apixaban, rivaroxaban, dabigatran, edoxaban) having replaced warfarin due to their superior safety and efficacy profile. However, for the subgroup of patients at very high risk for bleeding, alternative approaches need to be sought. LAAC is the most popular of these approaches, since the vast majority (>90%) of thrombi in nonvalvular AF originate in the LAA [144], which is a structure of variable shape and size with neurohormonal and reservoir functions. In AF, left atrial remodeling with alterations in shape, blood flow (stasis), and the presence of trabeculations are believed to be implicated in LAA thrombogenesis [145]. Surgical LAAC is a procedure with documented efficacy, as shown in the recently completed LAAOS-III randomized trial, as well as in a recent meta-analysis, for patients with AF undergoing cardiac surgery for another indication [146,147]. Percutaneous LAAC has also gained attention recently due to the safety and efficacy of the Watchman and Amplatzer devices (Table 2) [148]. Importantly, LAAC was noninferior to direct oral anticoagulants in terms of efficacy and safety in a randomized trial of high-risk patients [149]. Below, we review the current evidence in the field with the approved and under-investigation devices.

3.1. Percutaneous LAAC

3.1.1. Watchman and Watchman FLX

After the initial shortcoming of the PLAATO device [150], the Watchman device was the first approved. The landmark trial that compared the Watchman device to warfarin in nonvalvular AF with CHADS2 score ≥1 showed a lower rate of the primary endpoint (stroke, systemic embolism, and cardiovascular/unexplained death) after a 3.8-year follow-up with the device implantation [151]. The device’s safety and efficacy have been further supported by the evaluation of the Continued Access to PROTECT-AF and Continued Access to PREVAIL registries [152]. A recently reported analysis of the National Cardiovascular Data Registry LAAO Registry highlighted acceptable 1-year rates of ischemic stroke, mortality, and major bleeding [153]. Watchman implantation also led to quality-of-life improvements compared to warfarin [154]. Moreover, the device implantation led to ameliorated left atrial function, evidenced by increased ejection fraction and peak atrial contraction strain [155].
Regarding potential complications, the presence of peridevice leak ≥5 mm should be considered, however, since it may be associated with an increased risk of thromboembolism [156,157]. Device-related thrombosis is also an infrequent complication that may be accompanied by a higher risk of thrombotic events [158]. Risk factors for device-related thrombosis have been identified, namely hypercoagulability disorders, pericardial effusion, renal impairment, implantation depth >10 mm from the pulmonary vein limbus, and nonparoxysmal AF [159]. Device embolization into the aorta, left atrial cavity, and left ventricle are other catastrophic complications that can occur intraoperatively or postoperatively [160]. Embolized devices can be rescued percutaneously or through surgical retrieval, especially in cases of device embolization in the left ventricle, mitral apparatus, and descending aorta [160].
A second-generation device, the Watchman FLX, aiming at a simpler implantation with full recapture and repositioning, was evaluated in 165 elderly patients with a mean CHA2DS2-VASc score of 4.4 [161]. Importantly, the majority of patients had a history of major bleeding, and one quarter of LAAs were considered to have complex morphology. The rate of procedure-related complications was low, and no periprocedural major adverse events were noted. Only 6.7% received anticoagulants at discharge. The rates of peridevice leak and device-related thrombosis were low in the short term. Similar results were reported from a single European center in 91 patients [162]. Ultimately, the PINNACLE FLX trial established its high efficacy and safety [163], even in populations with previously failed Watchman 2.5 implantation or prohibitive LAA anatomy [164]. Compared with the original Watchman device, Watchman FLX had a similar short- and mid-term safety profile, with a superior sealing rate [165,166,167].

3.1.2. Amplatzer Cardiac Plug and Amulet

As early as 2003, the Amplatzer atrial septal occluder has been tried in patients with AF, demonstrating adequate efficacy and safety [168]. Early experiences with the Amplatzer cardiac plug (ACP) have been encouraging [169,170,171], and they were boosted by large registry data, showing high procedural success, low 1-year mortality and stroke/systemic thromboembolism rates, as well as few bleeding events [172]. At long-term follow-up, the ACP implantation was associated with lower ischemic stroke rates than those predicted by conventional risk scores [173]. Importantly, age and renal function should not be deterring factors, as the elderly and those with various degrees of renal dysfunction may exhibit similar efficacy and safety [174,175]. Adequate preoperative assessment with coronary-computed tomography angiography and follow-ups of patients with risk factors for device-related thrombosis (high ischemic risk scores, platelet count, and ejection fraction) need to be considered in these patients [176,177]. However, neither device-associated thrombosis nor peridevice leak were predictive of an increased risk of adverse cardiovascular events [178].
The next generation Amplatzer Amulet device, initially introduced in 2013, has a similar design to ACP, but offers easier implantation and reduced periprocedural complications. According to the results of a prospective global observational study including 1088 patients at high ischemic and bleeding risk who underwent its implantation, the rate of cardiovascular death or ischemic stroke was 8.7% at a 2-year follow-up [179]. This was accompanied by low rates of periprocedural major adverse events (4.0%), peridevice leak ≥3 mm (1.6%), and device-related thrombus (1.6%). Interestingly, a 67% reduction in the ischemic stroke rate compared to the predicted risk score was noted. Moreover, major bleeding event rates did not exceed 10%. The ischemic stroke risk reduction was more prominent in patients with CHA2DS2-VASc score ≥3 [180]. In addition, the results were similar across all age groups [181]. At 1-year follow-up, device-related thrombosis was encountered in 17 patients, and were associated with a greater risk of ischemic cerebrovascular events (HR 5.27, 95% CI 1.58017.55, p = 0.007), with a larger LAA orifice width being a predictive factor (HR 1.09, 95% CI 1.00–1.19, p = 0.04) [182].
When comparing the two Amplatzer LAAC devices, no major differences in the primary efficacy and safety endpoints were identified [183]. Interestingly, LAAC with Amplatzer devices led to a net clinical benefit compared with medical therapy in a propensity score-matched cohort study, by reducing the risk of stroke, systemic embolism, and cardiovascular/unexplained death (HR 0.70, 95% CI 0.53–0.95, p = 0.026) [184]. When the Amplatzer Amulet device was compared to direct oral anticoagulants, patients in the device group experienced fewer major bleeding events (HR 0.62, 95% CI 0.49–0.79), together with lower death rates (HR 0.53, 95% CI 0.43–0.64) [185]. Finally, although Amplatzer and Watchman devices appear to be similar in terms of efficacy and safety in previously reported registry data analyses [186,187], the lately published Amulet IDE randomized controlled trial pointed to a higher rate of periprocedural complications with the Amplatzer Amulet device compared to Watchman [188].

3.1.3. Other Devices

Perhaps the most extensively studied device other than Watchman and Amplatzer is the LARIAT system, combining endocardial and epicardial manipulation, ultimately ligating the LAA. Although early studies suggested an efficient LAA closure, this was hindered by high rates of procedural complications such as pericardial effusion, pericarditis, and major bleeding [189,190]. Periprocedural colchicine administration may be able to mitigate pericardial damage, as shown in two studies [191,192]. Among the interesting observations made with the LARIAT system are the reduction in AF burden [193], and the reversal of adverse atrial remodeling even in cases of incomplete closure [194,195,196]. Regarding major clinical endpoints, LARIAT was effective in reducing thromboembolic and bleeding events, as well as mortality rate, compared to a control group [197,198,199]. However, small leaks, which are not uncommon, should be considered, as they are predictive of adverse outcomes [200,201]. Sealing leaks with occluders may be a reasonable option in this scenario [202]. Compared to endocardial LAAC devices, the LARIAT system is associated with a higher short-term complication rate [203], but similar mid-term efficacy [204]. The second-generation LARIAT+ has improved features, such as snare expansion from 40 mm to 45 mm, the addition of a platinum–iridium ‘L’ Marker that allows identification of the device orientation under fluoroscopy, and a stainless steel wire braid on catheter shaft that provides improved ‘torque-ability’ of the catheter. Initial reports from registries highlight the high rates of acute and short-term LAAC, paired with low periprocedural complications and few thromboembolic events at longer term follow-up [205,206].
The LAmbre is a relatively new device that may be helpful in occluding large LAA, with its additional stabilization mechanism which catches the LAA trabeculations using its eight claws. The implantation success rate was 89.9% [207], with peridevice leak being detected in 17% at 1-year follow-up in one study [208]. This was not accompanied by a higher thromboembolic risk, however [208], perhaps due to the absence of significant leaks. Device-related thrombosis was rare, and no device embolization was documented [207]. Concerning mid-term outcomes, there was a low risk of death or ischemic cerebrovascular events, and major bleeding was seen in 11% of the patients at 1-year follow-up [209]. In a study with a mean follow-up of 37.8 months, the rates of death, stroke, and device-related thrombus were 7.1%, 3.6%, and 3.6%, respectively [210]. When compared to the Amplatzer and Watchman devices, the clinical efficacy and safety endpoints were met with similar frequency [211,212].

4. Conclusions

The milestones achieved in percutaneous approaches for atrial fibrillation, namely catheter ablation and left atrial appendage closure, constitute safe and effective treatment modalities in rhythm control and stroke prevention, respectively. Although currently reserved for a carefully selected group of patients, continuous research and persistently positive clinical trial data in a broad range of atrial fibrillation patients may mandate the need for the expansion of their indications.

Author Contributions

Conceptualization, P.T., Methodology, P.T., writing—original draft preparation, P.T., writing—review and editing, E.O., A.S.A., G.S., K.T. and D.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lippi, G.; Sanchis-Gomar, F.; Cervellin, G. Global epidemiology of atrial fibrillation: An increasing epidemic and public health challenge. Int. J. Stroke 2021, 16, 217–221. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, A.; Green, J.B.; Halperin, J.L.; Piccini, J.P. Atrial Fibrillation and Diabetes Mellitus: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2019, 74, 1107–1115. [Google Scholar] [CrossRef] [PubMed]
  3. Verdecchia, P.; Angeli, F.; Reboldi, G. Hypertension and Atrial Fibrillation: Doubts and Certainties From Basic and Clinical Studies. Circ. Res. 2018, 122, 352–368. [Google Scholar] [CrossRef] [PubMed]
  4. Javed, S.; Gupta, D.; Lip, G.Y.H. Obesity and atrial fibrillation: Making inroads through fat. Eur. Heart J. Cardiovasc. Pharmacother. 2021, 7, 59–67. [Google Scholar] [CrossRef]
  5. Sagris, M.; Vardas, E.P.; Theofilis, P.; Antonopoulos, A.S.; Oikonomou, E.; Tousoulis, D. Atrial Fibrillation: Pathogenesis, Predisposing Factors, and Genetics. Int. J. Mol. Sci. 2021, 23, 6. [Google Scholar] [CrossRef]
  6. Rydén, L.; Sacuiu, S.; Wetterberg, H.; Najar, J.; Guo, X.; Kern, S.; Zettergren, A.; Shams, S.; Pereira, J.B.; Wahlund, L.-O.; et al. Atrial Fibrillation, Stroke, and Silent Cerebrovascular Disease: A Population-based MRI Study. Neurology 2021, 97, e1608–e1619. [Google Scholar] [CrossRef]
  7. Carlisle, M.A.; Fudim, M.; DeVore, A.D.; Piccini, J.P. Heart Failure and Atrial Fibrillation, Like Fire and Fury. JACC: Heart Fail. 2019, 7, 447–456. [Google Scholar] [CrossRef]
  8. Madhavan, M.; Graff-Radford, J.; Piccini, J.P.; Gersh, B.J. Cognitive dysfunction in atrial fibrillation. Nat. Rev. Cardiol. 2018, 15, 744–756. [Google Scholar] [CrossRef]
  9. Randolph, T.C.; Simon, D.N.; Thomas, L.; Allen, L.A.; Fonarow, G.; Gersh, B.J.; Kowey, P.R.; Reiffel, J.A.; Naccarelli, G.V.; Chan, P.S.; et al. Patient factors associated with quality of life in atrial fibrillation. Am. Heart J. 2016, 182, 135–143. [Google Scholar] [CrossRef]
  10. Schnabel, R.B.; Michal, M.; Wilde, S.; Wiltink, J.; Wild, P.S.; Sinning, C.R.; Lubos, E.; Ojeda, F.M.; Zeller, T.; Munzel, T.; et al. Depression in Atrial Fibrillation in the General Population. PLoS ONE 2013, 8, e79109. [Google Scholar] [CrossRef] [Green Version]
  11. Freeman, J.V.; Wang, Y.; Akar, J.; Desai, N.; Krumholz, H. National Trends in Atrial Fibrillation Hospitalization, Readmission, and Mortality for Medicare Beneficiaries, 1999–2013. Circulation 2017, 135, 1227–1239. [Google Scholar] [CrossRef]
  12. Kirchhof, P.; Camm, A.J.; Goette, A.; Brandes, A.; Eckardt, L.; Elvan, A.; Fetsch, T.; van Gelder, I.C.; Haase, D.; Haegeli, L.M.; et al. Early Rhythm-Control Therapy in Patients with Atrial Fibrillation. N. Engl. J. Med. 2020, 383, 1305–1316. [Google Scholar] [CrossRef]
  13. Willems, S.; Borof, K.; Brandes, A.; Breithardt, G.; Camm, A.J.; Crijns, H.J.G.M.; Eckardt, L.; Gessler, N.; Goette, A.; Haegeli, L.M.; et al. Systematic, early rhythm control strategy for atrial fibrillation in patients with or without symptoms: The EAST-AFNET 4 trial. Eur. Heart J. 2022, 43, 1219–1230. [Google Scholar] [CrossRef]
  14. Haïssaguerre, M.; Jaïs, P.; Shah, D.C.; Takahashi, A.; Hocini, M.; Quiniou, G.; Garrigue, S.; Le Mouroux, A.; Le Métayer, P.; Clémenty, J. Spontaneous Initiation of Atrial Fibrillation by Ectopic Beats Originating in the Pulmonary Veins. N. Engl. J. Med. 1998, 339, 659–666. [Google Scholar] [CrossRef]
  15. Pappone, C.; Oreto, G.; Lamberti, F.; Vicedomini, G.; Loricchio, M.L.; Shpun, S.; Rillo, M.; Calabrò, M.P.; Conversano, A.; Ben-Haim, S.A.; et al. Catheter Ablation of Paroxysmal Atrial Fibrillation Using a 3D Mapping System. Circulation 1999, 100, 1203–1208. [Google Scholar] [CrossRef]
  16. Arentz, T.; Weber, R.; Bürkle, G.; Herrera, C.; Blum, T.; Stockinger, J.; Minners, J.; Neumann, F.J.; Kalusche, D. Small or Large Isolation Areas Around the Pulmonary Veins for the Treatment of Atrial Fibrillation? Results from a prospective randomized study. Circulation 2007, 115, 3057–3063. [Google Scholar] [CrossRef]
  17. Proietti, R.; Santangeli, P.; Di Biase, L.; Joza, J.; Bernier, M.L.; Wang, Y.; Sagone, A.; Viecca, M.; Essebag, V.; Natale, A. Comparative Effectiveness of Wide Antral Versus Ostial Pulmonary Vein Isolation: A systematic review and meta-analysis. Circ. Arrhythmia Electrophysiol. 2014, 7, 39–45. [Google Scholar] [CrossRef]
  18. Redfearn, D.P.; Skanes, A.C.; Gula, L.J.; Griffith, M.J.; Marshall, H.J.; Stafford, P.J.; Krahn, A.D.; Yee, R.; Klein, G.J. Noninvasive Assessment of Atrial Substrate Change after Wide Area Circumferential Ablation: A Comparison with Segmental Pulmonary Vein Isolation. Ann. Noninvasive Electrocardiol. 2007, 12, 329–337. [Google Scholar] [CrossRef]
  19. Calkins, H.; Hindricks, G.; Cappato, R.; Kim, Y.-H.; Saad, E.B.; Aguinaga, L.; Akar, J.G.; Badhwar, V.; Brugada, J.; Camm, J.; et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation. Heart Rhythm. 2017, 14, e275–e444. [Google Scholar] [CrossRef]
  20. Kino, T.; Kagimoto, M.; Yamada, T.; Ishii, S.; Asai, M.; Asano, S.; Yano, H.; Ishikawa, T.; Ishigami, T. Optimal Anticoagulant Strategy for Periprocedural Management of Atrial Fibrillation Ablation: A Systematic Review and Network Meta-Analysis. J. Clin. Med. 2022, 11, 1872. [Google Scholar] [CrossRef]
  21. Xu, J.; Gao, Y.; Liu, C.; Wang, Y. Radiofrequency ablation for treatment of atrial fibrillation with the use of intracardiac echocardiography versus without intracardiac echocardiography: A meta-analysis of observational and randomized studies. J. Cardiovasc. Electrophysiol. 2022, 33, 897–907. [Google Scholar] [CrossRef]
  22. Song, J.; Zhang, Q.; Ye, L.; Zheng, Y.; Wang, L. The comparison of catheter ablation on hard outcomes versus medical treatment for atrial fibrillation patients: A meta-analysis of randomized, controlled trials with trial sequential analysis. PLoS ONE 2022, 17, e0262702. [Google Scholar] [CrossRef]
  23. Leung, L.W.M.; Imhoff, R.J.; Marshall, H.J.; Frame, D.; Mallow, P.J.; Goldstein, L.; Wei, T.; Velleca, M.; Taylor, H.; Gallagher, M.M. Cost-effectiveness of catheter ablation versus medical therapy for the treatment of atrial fibrillation in the United Kingdom. J. Cardiovasc. Electrophysiol. 2022, 33, 164–175. [Google Scholar] [CrossRef]
  24. Packer, D.L.; Mark, D.B.; Robb, R.A.; Monahan, K.H.; Bahnson, T.D.; Poole, J.E.; Noseworthy, P.A.; Rosenberg, Y.D.; Jeffries, N.; Mitchell, L.B.; et al. Effect of Catheter Ablation vs Antiarrhythmic Drug Therapy on Mortality, Stroke, Bleeding, and Cardiac Arrest Among Patients With Atrial Fibrillation: The CABANA Randomized Clinical Trial. JAMA 2019, 321, 1261–1274. [Google Scholar] [CrossRef]
  25. Bahnson, T.D.; Giczewska, A.; Mark, D.B.; Russo, A.M.; Monahan, K.H.; Al-Khalidi, H.R.; Silverstein, A.P.; Poole, J.E.; Lee, K.L.; Packer, D.L.; et al. Association Between Age and Outcomes of Catheter Ablation Versus Medical Therapy for Atrial Fibrillation: Results From the CABANA Trial. Circulation 2022, 145, 796–804. [Google Scholar] [CrossRef]
  26. Poole, J.E.; Bahnson, T.D.; Monahan, K.H.; Johnson, G.; Rostami, H.; Silverstein, A.P.; Al-Khalidi, H.R.; Rosenberg, Y.; Mark, D.B.; Lee, K.L.; et al. Recurrence of Atrial Fibrillation After Catheter Ablation or Antiarrhythmic Drug Therapy in the CABANA Trial. J. Am. Coll. Cardiol. 2020, 75, 3105–3118. [Google Scholar] [CrossRef]
  27. Lee, W.; Wu, P.; Fang, C.; Chen, H.; Chen, M. Impact of chronic kidney disease on atrial fibrillation recurrence following radiofrequency and cryoballoon ablation: A meta-analysis. Int. J. Clin. Pract. 2021, 75, e14173. [Google Scholar] [CrossRef]
  28. Garvanski, I.; Simova, I.; Angelkov, L.; Matveev, M. Predictors of Recurrence of AF in Patients After Radiofrequency Ablation. Eur. Cardiol. Rev. 2019, 14, 165–168. [Google Scholar] [CrossRef]
  29. Du, H.; Yang, L.; Hu, Z.; Zhang, H. Anxiety is associated with higher recurrence of atrial fibrillation after catheter ablation: A meta-analysis. Clin. Cardiol. 2022, 45, 243–250. [Google Scholar] [CrossRef]
  30. Rettmann, M.E.; Holmes, D.R., 3rd; Monahan, K.H.; Breen, J.F.; Bahnson, T.D.; Mark, D.B.; Poole, J.E.; Ellis, A.M.; Silverstein, A.P.; Al-Khalidi, H.R.; et al. Treatment-Related Changes in Left Atrial Structure in Atrial Fibrillation: Findings From the CABANA Imaging Substudy. Circ. Arrhythmia Electrophysiol. 2021, 14, e008540. [Google Scholar] [CrossRef]
  31. Boyalla, V.; Harling, L.; Snell, A.; Kralj-Hans, I.; Barradas-Pires, A.; Haldar, S.; Khan, H.R.; Cleland, J.G.F.; Athanasiou, T.; Harding, S.E.; et al. Biomarkers as predictors of recurrence of atrial fibrillation post ablation: An updated and expanded systematic review and meta-analysis. Clin. Res. Cardiol. 2022, 111, 680–691. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, G.; Wu, Y. Circulating Galectin-3 and Atrial Fibrillation Recurrence after Catheter Ablation: A Meta-Analysis. Cardiovasc. Ther. 2019, 2019, 4148129. [Google Scholar] [CrossRef] [PubMed]
  33. Hindricks, G.; Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.J.; Blomström-Lundqvist, C.; Boriani, G.; Castella, M.; Dan, G.-A.; Dilaveris, P.E.; et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J. 2021, 42, 373–498. [Google Scholar] [CrossRef] [PubMed]
  34. Turagam, M.K.; Musikantow, D.; Whang, W.; Koruth, J.S.; Miller, M.A.; Langan, M.-N.; Sofi, A.; Choudry, S.; Dukkipati, S.R.; Reddy, V.Y. Assessment of Catheter Ablation or Antiarrhythmic Drugs for First-line Therapy of Atrial Fibrillation: A Meta-analysis of Randomized Clinical Trials. JAMA Cardiol. 2021, 6, 697–705. [Google Scholar] [CrossRef]
  35. Saglietto, A.; Ballatore, A.; Xhakupi, H.; De Ferrari, G.M.; Anselmino, M. Association of Catheter Ablation and Reduced Incidence of Dementia among Patients with Atrial Fibrillation during Long-Term Follow-Up: A Systematic Review and Meta-Analysis of Observational Studies. J. Cardiovasc. Dev. Dis. 2022, 9, 140. [Google Scholar] [CrossRef] [PubMed]
  36. Kuck, K.-H.; Lebedev, D.S.; Mikhaylov, E.N.; Romanov, A.; Gellér, L.; Kalējs, O.; Neumann, T.; Davtyan, K.; On, Y.K.; Popov, S.; et al. Catheter ablation or medical therapy to delay progression of atrial fibrillation: The randomized controlled atrial fibrillation progression trial (ATTEST). Europace 2021, 23, 362–369a. [Google Scholar] [CrossRef] [PubMed]
  37. de Vos, C.B.; Pisters, R.; Nieuwlaat, R.; Prins, M.H.; Tieleman, R.G.; Coelen, R.-J.S.; Heijkant, A.C.V.D.; Allessie, M.A.; Crijns, H.J. Progression From Paroxysmal to Persistent Atrial Fibrillation: Clinical Correlates and Prognosis. J. Am. Coll. Cardiol. 2010, 55, 725–731. [Google Scholar] [CrossRef]
  38. Tang, P.-T.; Davies, M.; Bashir, Y.; Betts, T.R.; Pedersen, M.; Rajappan, K.; Ginks, M.R.; Wijesurendra, R.S. Efficacy and safety of same-day discharge after atrial fibrillation ablation compared with post-procedural overnight stay: A systematic review and meta-analysis. Europace 2022, euac068. [Google Scholar] [CrossRef]
  39. Ejima, K.; Kato, K.; Okada, A.; Wakisaka, O.; Kimura, R.; Ishizawa, M.; Imai, T.; Toyama, Y.; Shoda, M.; Hagiwara, N. Comparison Between Contact Force Monitoring and Unipolar Signal Modification as a Guide for Catheter Ablation of Atrial Fibrillation: Prospective Multi-Center Randomized Study. Circ. Arrhythmia Electrophysiol. 2019, 12, e007311. [Google Scholar] [CrossRef]
  40. Kim, T.-H.; Park, J.; Uhm, J.-S.; Joung, B.; Lee, M.-H.; Pak, H.-N. Pulmonary vein reconnection predicts good clinical outcome after second catheter ablation for atrial fibrillation. Europace 2017, 19, 961–967. [Google Scholar] [CrossRef]
  41. Papageorgiou, N.; Karim, N.; Williams, J.; Garcia, J.; Creta, A.; Ang, R.; Srinivasan, N.; Providencia, R.; Hunter, R.J.; Dhinoja, M.; et al. Initial experience of the High-Density Grid catheter in patients undergoing catheter ablation for atrial fibrillation. J. Interv. Card. Electrophysiol. 2022, 63, 259–266. [Google Scholar] [CrossRef]
  42. Das, M.; Loveday, J.J.; Wynn, G.J.; Gomes, S.; Saeed, Y.; Bonnett, L.J.; Waktare, J.E.; Todd, D.M.; Hall, M.C.; Snowdon, R.L.; et al. Ablation index, a novel marker of ablation lesion quality: Prediction of pulmonary vein reconnection at repeat electrophysiology study and regional differences in target values. Europace 2017, 19, 775–783. [Google Scholar] [CrossRef]
  43. Duytschaever, M.; De Pooter, J.; Demolder, A.; El Haddad, M.; Phlips, T.; Strisciuglio, T.; Debonnaire, P.; Wolf, M.; Vandekerckhove, Y.; Knecht, S.; et al. Long-term impact of catheter ablation on arrhythmia burden in low-risk patients with paroxysmal atrial fibrillation: The CLOSE to CURE study. Heart Rhythm 2020, 17, 535–543. [Google Scholar] [CrossRef]
  44. Prasad, K.V.; Bonso, A.; Woods, C.E.; Goya, M.; Matsuo, S.; Padanilam, B.J.; Kreis, I.; Yang, F.; Williams, C.G.; Tranter, J.H.; et al. Lesion Index–guided workflow for the treatment of paroxysmal atrial fibrillation is safe and effective—Final results from the LSI Workflow Study. Heart Rhythm O2, 2022; in press. [Google Scholar] [CrossRef]
  45. Kim, D.; Yu, H.T.; Kim, T.-H.; Uhm, J.-S.; Joung, B.; Lee, M.-H.; Pak, H.-N. Electrical Posterior Box Isolation in Repeat Ablation for Atrial Fibrillation: A Prospective Randomized Clinical Study. JACC Clin. Electrophysiol. 2022, 8, 582–592. [Google Scholar] [CrossRef]
  46. Chieng, D.; Sugumar, H.; Ling, L.-H.; Segan, L.; Azzopardi, S.; Prabhu, S.; Al-Kaisey, A.; Voskoboinik, A.; Parameswaran, R.; Morton, J.B.; et al. Catheter ablation for persistent atrial fibrillation: A multicenter randomized trial of pulmonary vein isolation (PVI) versus PVI with posterior left atrial wall isolation (PWI)—The CAPLA study. Am. Heart J. 2022, 243, 210–220. [Google Scholar] [CrossRef]
  47. Aryana, A.; Pujara, D.K.; Allen, S.L.; Baker, J.H.; Espinosa, M.A.; Buch, E.F.; Srivatsa, U.; Ellis, E.; Makati, K.; Kowalski, M.; et al. Left atrial posterior wall isolation in conjunction with pulmonary vein isolation using cryoballoon for treatment of persistent atrial fibrillation (PIVoTAL): Study rationale and design. J. Interv. Card. Electrophysiol. 2021, 62, 187–198. [Google Scholar] [CrossRef]
  48. Kim, M.-Y.; Coyle, C.; Tomlinson, D.R.; Sikkel, M.B.; Sohaib, A.; Luther, V.; Leong, K.M.; Malcolme-Lawes, L.; Low, B.; Sandler, B.; et al. Ectopy-triggering ganglionated plexuses ablation to prevent atrial fibrillation: GANGLIA-AF study. Heart Rhythm 2022, 19, 516–524. [Google Scholar] [CrossRef]
  49. Rackley, J.; Nudy, M.; Gonzalez, M.D.; Naccarelli, G.; Maheshwari, A. Pulmonary vein isolation with adjunctive left atrial ganglionic plexus ablation for treatment of atrial fibrillation: A meta-analysis of randomized controlled trials. J. Interv. Card. Electrophysiol. 2022, 1–10. [Google Scholar] [CrossRef]
  50. Li, F.; Sun, J.-Y.; Wu, L.-D.; Zhang, L.; Qu, Q.; Wang, C.; Qian, L.-L.; Wang, R.-X. The Long-Term Outcomes of Ablation With Vein of Marshall Ethanol Infusion vs. Ablation Alone in Patients With Atrial Fibrillation: A Meta-Analysis. Front. Cardiovasc. Med. 2022, 9, 871654. [Google Scholar] [CrossRef]
  51. Leshem, E.; Zilberman, I.; Tschabrunn, C.M.; Barkagan, M.; Contreras-Valdes, F.M.; Govari, A.; Anter, E. High-Power and Short-Duration Ablation for Pulmonary Vein Isolation. JACC Clin. Electrophysiol. 2018, 4, 467–479. [Google Scholar] [CrossRef] [PubMed]
  52. Nilsson, B.; Chen, X.; Pehrson, S.; Svendsen, J.H. The effectiveness of a high output/short duration radiofrequency current application technique in segmental pulmonary vein isolation for atrial fibrillation. Europace 2006, 8, 962–965. [Google Scholar] [CrossRef] [PubMed]
  53. Shin, D.G.; Ahn, J.; Han, S.-J.; Lim, H.E. Efficacy of high-power and short-duration ablation in patients with atrial fibrillation: A prospective randomized controlled trial. EP Eurospace 2020, 22, 1495–1501. [Google Scholar] [CrossRef] [PubMed]
  54. Yavin, H.D.; Leshem, E.; Shapira-Daniels, A.; Sroubek, J.; Barkagan, M.; Haffajee, C.I.; Cooper, J.M.; Anter, E. Impact of High-Power Short-Duration Radiofrequency Ablation on Long-Term Lesion Durability for Atrial Fibrillation Ablation. JACC Clin. Electrophysiol. 2020, 6, 973–985. [Google Scholar] [CrossRef] [PubMed]
  55. Francke, A.; Scharfe, F.; Schoen, S.; Wunderlich, C.; Christoph, M. Reconnection patterns after CLOSE-guided 50 W high-power-short-duration circumferential pulmonary vein isolation and substrate modification—PV reconnection might no longer be an issue. J. Cardiovasc. Electrophysiol. 2022, 33, 1136–1145. [Google Scholar] [CrossRef] [PubMed]
  56. Kumagai, K.; Toyama, H. High-power, short-duration ablation during Box isolation for atrial fibrillation. J. Arrhythmia 2020, 36, 899–904. [Google Scholar] [CrossRef]
  57. Kusa, S.; Hachiya, H.; Sato, Y.; Hara, S.; Ohya, H.; Miwa, N.; Yamao, K.; Iesaka, Y.; Sasano, T. Superior vena cava isolation with 50 W high power, short duration ablation strategy. J. Cardiovasc. Electrophysiol. 2021, 32, 1602–1609. [Google Scholar] [CrossRef]
  58. Junarta, J.; Dikdan, S.J.; Upadhyay, N.; Bodempudi, S.; Shvili, M.Y.; Frisch, D.R. High-power short-duration versus standard-power standard-duration settings for repeat atrial fibrillation ablation. Heart Vessel. 2022, 37, 1003–1009. [Google Scholar] [CrossRef]
  59. Ravi, V.; Poudyal, A.; Abid, Q.-U.; Larsen, T.; Krishnan, K.; Sharma, P.S.; Trohman, R.G.; Huang, H.D. High-power short duration vs. conventional radiofrequency ablation of atrial fibrillation: A systematic review and meta-analysis. Europace 2021, 23, 710–721. [Google Scholar] [CrossRef]
  60. Kaneshiro, T.; Kamioka, M.; Hijioka, N.; Yamada, S.; Yokokawa, T.; Misaka, T.; Hikichi, T.; Yoshihisa, A.; Takeishi, Y. Characteristics of Esophageal Injury in Ablation of Atrial Fibrillation Using a High-Power Short-Duration Setting. Circ. Arrhythmia. Electrophysiol. 2020, 13, e008602. [Google Scholar] [CrossRef]
  61. Ayoub, T.; El Hajjar, A.H.; Sidhu, G.D.S.; Bhatnagar, A.; Zhang, Y.; Mekhael, M.; Noujaim, C.; Dagher, L.; Pottle, C.; Marrouche, N. Esophageal temperature during atrial fibrillation ablation poorly predicts esophageal injury: An observational study. Heart Rhythm O2 2021, 2, 570–577. [Google Scholar] [CrossRef]
  62. Müller, J.; Berkovitz, A.; Halbfass, P.; Nentwich, K.; Ene, E.; Sonne, K.; Simu, G.; Chakarov, I.; Barth, S.; Waechter, C.; et al. Acute oesophageal safety of high-power short duration with 50 W for atrial fibrillation ablation. Europace 2022, 24, 928–937. [Google Scholar] [CrossRef]
  63. Chen, S.; Chun, K.J.; Tohoku, S.; Bordignon, S.; Urbanek, L.; Willems, F.; Plank, K.; Hilbert, M.; Konstantinou, A.; Tsianakas, N.; et al. Esophageal Endoscopy After Catheter Ablation of Atrial Fibrillation Using Ablation-Index Guided High-Power: Frankfurt AI-HP ESO-I. JACC Clin. Electrophysiol. 2020, 6, 1253–1261. [Google Scholar] [CrossRef]
  64. Francke, A.; Taha, N.S.; Scharfe, F.; Schoen, S.; Wunderlich, C.; Christoph, M. Procedural efficacy and safety of standardized, ablation index guided fixed 50 W high-power short-duration pulmonary vein isolation and substrate modification using the CLOSE protocol. J. Cardiovasc. Electrophysiol. 2021, 32, 2408–2417. [Google Scholar] [CrossRef]
  65. Reddy, V.Y.; Grimaldi, M.; De Potter, T.; Vijgen, J.M.; Bulava, A.; Duytschaever, M.F.; Martinek, M.; Natale, A.; Knecht, S.; Neuzil, P.; et al. Pulmonary Vein Isolation With Very High Power, Short Duration, Temperature-Controlled Lesions: The QDOT-FAST Trial. JACC Clin. Electrophysiol. 2019, 5, 778–786. [Google Scholar] [CrossRef]
  66. Tilz, R.R.; Sano, M.; Vogler, J.; Fink, T.; Saraei, R.; Sciacca, V.; Kirstein, B.; Phan, H.-L.; Hatahet, S.; Lopez, L.D.; et al. Very high-power short-duration temperature-controlled ablation versus conventional power-controlled ablation for pulmonary vein isolation: The fast and furious—AF study. IJC Heart Vasc. 2021, 35, 100847. [Google Scholar] [CrossRef]
  67. Kottmaier, M.; Popa, M.-A.; Bourier, F.; Reents, T.; Cifuentes, J.; Semmler, V.; Telishevska, M.; Otgonbayar, U.; Koch-Büttner, K.; Lennerz, C.; et al. Safety and outcome of very high-power short-duration ablation using 70 W for pulmonary vein isolation in patients with paroxysmal atrial fibrillation. Europace 2020, 22, 388–393. [Google Scholar] [CrossRef]
  68. Halbfass, P.; Wielandts, J.-Y.; Knecht, S.; Waroux, J.-B.L.P.D.; Tavernier, R.; De Wilde, V.; Sonne, K.; Nentwich, K.; Ene, E.; Berkovitz, A.; et al. Safety of very high-power short-duration radiofrequency ablation for pulmonary vein isolation: A two-centre report with emphasis on silent oesophageal injury. EP Eurospace 2021, 24, 400–405. [Google Scholar] [CrossRef]
  69. Velagić, V.; de Asmundis, C.; Mugnai, G.; Hünük, B.; Hacioğlu, E.; Ströker, E.; Moran, D.; Ruggiero, D.; Poelaert, J.; Verborgh, C.; et al. Learning curve using the second-generation cryoballoon ablation. J. Cardiovasc. Med. 2017, 18, 518–527. [Google Scholar] [CrossRef]
  70. Osório, T.G.; Coutiño, H.-E.; Brugada, P.; Chierchia, G.-B.; De Asmundis, C. Recent advances in cryoballoon ablation for atrial fibrillation. Expert Rev. Med. Devices 2019, 16, 799–808. [Google Scholar] [CrossRef]
  71. Andrade, J.G.; Wells, G.A.; Deyell, M.W.; Bennett, M.; Essebag, V.; Champagne, J.; Roux, J.-F.; Yung, D.; Skanes, A.; Khaykin, Y.; et al. Cryoablation or Drug Therapy for Initial Treatment of Atrial Fibrillation. N. Engl. J. Med. 2021, 384, 305–315. [Google Scholar] [CrossRef]
  72. Kuniss, M.; Pavlovic, N.; Velagic, V.; Hermida, J.S.; Healey, S.; Arena, G.; Badenco, N.; Meyer, C.; Chen, J.; Iacopino, S.; et al. Cryoballoon ablation vs. antiarrhythmic drugs: First-line therapy for patients with paroxysmal atrial fibrillation. Europace 2021, 23, 1033–1041. [Google Scholar] [CrossRef]
  73. Wazni, O.M.; Dandamudi, G.; Sood, N.; Hoyt, R.; Tyler, J.; Durrani, S.; Niebauer, M.; Makati, K.; Halperin, B.; Gauri, A.; et al. Cryoballoon Ablation as Initial Therapy for Atrial Fibrillation. N. Engl. J. Med. 2021, 384, 316–324. [Google Scholar] [CrossRef]
  74. Wazni, O.; Dandamudi, G.; Sood, N.; Hoyt, R.; Tyler, J.; Durrani, S.; Niebauer, M.; Makati, K.; Halperin, B.; Gauri, A.; et al. Quality of life after the initial treatment of atrial fibrillation with cryoablation versus drug therapy. Heart Rhythm 2022, 19, 197–205. [Google Scholar] [CrossRef]
  75. Pavlovic, N.; Chierchia, G.-B.; Velagic, V.; Hermida, J.S.; Healey, S.; Arena, G.; Badenco, N.; Meyer, C.; Chen, J.; Iacopino, S.; et al. Initial rhythm control with cryoballoon ablation vs drug therapy: Impact on quality of life and symptoms. Am. Heart J. 2021, 242, 103–114. [Google Scholar] [CrossRef]
  76. Vrachatis, D.A.; Papathanasiou, K.A.; Kossyvakis, C.; Kazantzis, D.; Giotaki, S.G.; Deftereos, G.; Sanz-Sánchez, J.; Raisakis, K.; Kaoukis, A.; Avramides, D.; et al. Early arrhythmia recurrence after cryoballoon ablation in atrial fibrillation: A systematic review and meta-analysis. J. Cardiovasc. Electrophysiol. 2022, 33, 527–539. [Google Scholar] [CrossRef]
  77. Andrade, J.G.; Champagne, J.; Dubuc, M.; Deyell, M.W.; Verma, A.; Macle, L.; Leong-Sit, P.; Novak, P.; Badra-Verdu, M.; Sapp, J.; et al. Cryoballoon or Radiofrequency Ablation for Atrial Fibrillation Assessed by Continuous Monitoring: A Randomized Clinical Trial. Circulation 2019, 140, 1779–1788. [Google Scholar] [CrossRef]
  78. Pak, H.-N.; Park, J.-W.; Yang, S.-Y.; Kim, T.-H.; Uhm, J.-S.; Joung, B.; Lee, M.-H.; Yu, H.T. Cryoballoon Versus High-Power, Short-Duration Radiofrequency Ablation for Pulmonary Vein Isolation in Patients With Paroxysmal Atrial Fibrillation: A Single-Center, Prospective, Randomized Study. Circ. Arrhythmia Electrophysiol. 2021, 14, e010040. [Google Scholar] [CrossRef]
  79. Lavallaz, J.D.F.D.; Badertscher, P.; Kobori, A.; Kuck, K.-H.; Brugada, J.; Boveda, S.; Providência, R.; Khoueiry, Z.; Luik, A.; Squara, F.; et al. Sex-specific efficacy and safety of cryoballoon versus radiofrequency ablation for atrial fibrillation: An individual patient data meta-analysis. Heart Rhythm 2020, 17, 1232–1240. [Google Scholar] [CrossRef]
  80. Chun, J.K.; Bordignon, S.; Last, J.; Mayer, L.; Tohoku, S.; Zanchi, S.; Bianchini, L.; Bologna, F.; Nagase, T.; Urbanek, L.; et al. Cryoballoon Versus Laserballoon: Insights From the First Prospective Randomized Balloon Trial in Catheter Ablation of Atrial Fibrillation. Circ. Arrhythmia Electrophysiol. 2021, 14, e009294. [Google Scholar] [CrossRef]
  81. Schmidt, B.; Neuzil, P.; Luik, A.; Osca Asensi, J.; Schrickel, J.W.; Deneke, T.; Bordignon, S.; Petru, J.; Merkel, M.; Sediva, L.; et al. Laser Balloon or Wide-Area Circumferential Irrigated Radiofrequency Ablation for Persistent Atrial Fibrillation: A Multicenter Prospective Randomized Study. Circ. Arrhythm. Electrophysiol. 2017, 10, e005767. [Google Scholar] [CrossRef] [PubMed]
  82. Schiavone, M.; Gasperetti, A.; Montemerlo, E.; Pozzi, M.; Sabato, F.; Piazzi, E.; Ruggiero, D.; De Ceglia, S.; Viecca, M.; Calkins, H.; et al. Long-term comparisons of atrial fibrillation ablation outcomes with a cryoballoon or laser-balloon: A propensity-matched analysis based on continuous rhythm monitoring. Hell. J. Cardiol. 2022, 65, 1–7. [Google Scholar] [CrossRef] [PubMed]
  83. Üçer, E.; Fredersdorf, S.; Seegers, J.; Poschenrieder, F.; Hauck, C.; Maier, L.; Jungbauer, C. High Predictive Value of Adenosine Provocation in Predicting Atrial Fibrillation Recurrence After Pulmonary Vein Isolation With Visually Guided Laser Balloon Compared With Radiofrequency Ablation. Circ. J. 2020, 84, 404–410. [Google Scholar] [CrossRef] [PubMed]
  84. Figueras i Ventura, R.M.; Mǎrgulescu, A.D.; Benito, E.M.; Alarcón, F.; Enomoto, N.; Prat-Gonzalez, S.; Perea, R.J.; Borràs, R.; Chipa, F.; Arbelo, E.; et al. Postprocedural LGE-CMR comparison of laser and radiofrequency ablation lesions after pulmonary vein isolation. J. Cardiovasc. Electrophysiol. 2018, 29, 1065–1072. [Google Scholar] [CrossRef]
  85. Skeete, J.; Sharma, P.S.; Kenigsberg, D.; Pietrasik, G.; Osman, A.F.; Ravi, V.; Du-Fay-De-Lavallaz, J.M.; Post, Z.; Wasserlauf, J.; Larsen, T.R.; et al. Wide area circumferential ablation for pulmonary vein isolation using radiofrequency versus laser balloon ablation. J. Arrhythmia 2022, 38, 336–345. [Google Scholar] [CrossRef]
  86. Sasaki, Y.; Kobori, A.; Ishikura, M.; Murai, R.; Okada, T.; Toyota, T.; Taniguchi, T.; Kim, K.; Ehara, N.; Kinoshita, M.; et al. Effectiveness of a manual dragging laser irradiation technique using the first-generation endoscopic laser balloon ablation system for pulmonary vein isolation. J. Arrhythmia 2022, 38, 327–335. [Google Scholar] [CrossRef]
  87. Guenancia, C.; Hammache, N.; Docq, C.; Benali, K.; Hooks, D.; Echivard, M.; Pace, N.; Magnin-Poull, I.; de Chillou, C.; Sellal, J.-M. Efficacy and Safety of Second and Third-Generation Laser Balloon for Paroxysmal Atrial Fibrillation Ablation Compared to Radiofrequency Ablation: A Matched-Cohort. J. Cardiovasc. Dev. Dis. 2021, 8, 183. [Google Scholar] [CrossRef]
  88. Tohoku, S.; Bordignon, S.; Chen, S.; Bologna, F.; Urbanek, L.; Operhalski, F.; Chun, K.J.; Schmidt, B. Validation of lesion durability following pulmonary vein isolation using the new third-generation laser balloon catheter in patients with recurrent atrial fibrillation. J. Cardiol. 2021, 78, 388–396. [Google Scholar] [CrossRef]
  89. Nakatani, Y.; Sridi-Cheniti, S.; Cheniti, G.; Ramirez, F.D.; Goujeau, C.; André, C.; Nakashima, T.; Eggert, C.; Schneider, C.; Viswanathan, R.; et al. Pulsed field ablation prevents chronic atrial fibrotic changes and restrictive mechanics after catheter ablation for atrial fibrillation. Europace 2021, 23, 1767–1776. [Google Scholar] [CrossRef]
  90. Kuroki, K.; Whang, W.; Eggert, C.; Lam, J.; Leavitt, J.; Kawamura, I.; Reddy, A.; Morrow, B.; Schneider, C.; Petru, J.; et al. Ostial dimensional changes after pulmonary vein isolation: Pulsed field ablation vs radiofrequency ablation. Heart Rhythm 2020, 17, 1528–1535. [Google Scholar] [CrossRef]
  91. Stewart, M.T.; Haines, D.E.; Verma, A.; Kirchhof, N.; Barka, N.; Grassl, E.; Howard, B. Intracardiac pulsed field ablation: Proof of feasibility in a chronic porcine model. Heart Rhythm 2019, 16, 754–764. [Google Scholar] [CrossRef]
  92. Koruth, J.; Kuroki, K.; Iwasawa, J.; Enomoto, Y.; Viswanathan, R.; Brose, R.; Buck, E.D.; Speltz, M.; Dukkipati, S.R.; Reddy, V. Preclinical Evaluation of Pulsed Field Ablation: Electrophysiological and Histological Assessment of Thoracic Vein Isolation. Circ. Arrhythmia Electrophysiol. 2019, 12, e007781. [Google Scholar] [CrossRef]
  93. Reddy, V.Y.; Koruth, J.; Jais, P.; Petru, J.; Timko, F.; Skalsky, I.; Hebeler, R.; Labrousse, L.; Barandon, L.; Kralovec, S.; et al. Ablation of Atrial Fibrillation With Pulsed Electric Fields: An Ultra-Rapid, Tissue-Selective Modality for Cardiac Ablation. JACC Clin. Electrophysiol. 2018, 4, 987–995. [Google Scholar] [CrossRef]
  94. Reddy, V.Y.; Neuzil, P.; Koruth, J.S.; Petru, J.; Funosako, M.; Cochet, H.; Sediva, L.; Chovanec, M.; Dukkipati, S.R.; Jais, P. Pulsed Field Ablation for Pulmonary Vein Isolation in Atrial Fibrillation. J. Am. Coll. Cardiol. 2019, 74, 315–326. [Google Scholar] [CrossRef]
  95. Ekanem, E.; Reddy, V.Y.; Schmidt, B.; Reichlin, T.; Neven, K.; Metzner, A.; Hansen, J.; Blaauw, Y.; Maury, P.; Arentz, T.; et al. Multi-national survey on the methods, efficacy, and safety on the post-approval clinical use of pulsed field ablation (MANIFEST-PF). Europace 2022, 24, 1256–1266. [Google Scholar] [CrossRef]
  96. Bohnen, M.; Weber, R.; Minners, J.; Jadidi, A.; Eichenlaub, M.; Neumann, F.J.; Arentz, T.; Lehrmann, H. Characterization of circumferential antral pulmonary vein isolation areas resulting from pulsed-field catheter ablation. Europace 2022, euac111. [Google Scholar] [CrossRef]
  97. Verma, A.; Boersma, L.; Haines, D.E.; Natale, A.; Marchlinski, F.E.; Sanders, P.; Calkins, H.; Packer, D.L.; Hummel, J.; Onal, B.; et al. First-in-Human Experience and Acute Procedural Outcomes Using a Novel Pulsed Field Ablation System: The PULSED AF Pilot Trial. Circ. Arrhythmia Electrophysiol. 2022, 15, e010168. [Google Scholar] [CrossRef]
  98. Reddy, V.Y.; Anter, E.; Rackauskas, G.; Peichl, P.; Koruth, J.S.; Petru, J.; Funasako, M.; Minami, K.; Natale, A.; Jaïs, P.; et al. Lattice-Tip Focal Ablation Catheter That Toggles Between Radiofrequency and Pulsed Field Energy to Treat Atrial Fibrillation: A First-in-Human Trial. Circ. Arrhythmia Electrophysiol. 2020, 13, e008718. [Google Scholar] [CrossRef]
  99. Kueffer, T.; Baldinger, S.H.; Servatius, H.; Madaffari, A.; Seiler, J.; Muhl, A.; Franzeck, F.; Thalmann, G.; Asatryan, B.; Haeberlin, A.; et al. Validation of a multipolar pulsed-field ablation catheter for endpoint assessment in pulmonary vein isolation procedures. Europace 2022, 24, 1248–1255. [Google Scholar] [CrossRef]
  100. Sohara, H.; Takeda, H.; Ueno, H.; Oda, T.; Satake, S. Feasibility of the Radiofrequency Hot Balloon Catheter for Isolation of the Posterior Left Atrium and Pulmonary Veins for the Treatment of Atrial Fibrillation. Circ. Arrhythmia Electrophysiol. 2009, 2, 225–232. [Google Scholar] [CrossRef] [Green Version]
  101. Yamaguchi, Y.; Sohara, H.; Takeda, H.; Nakamura, Y.; Ihara, M.; Higuchi, S.; Satake, S. Long-Term Results of Radiofrequency Hot Balloon Ablation in Patients With Paroxysmal Atrial Fibrillation: Safety and Rhythm Outcomes. J. Cardiovasc. Electrophysiol. 2015, 26, 1298–1306. [Google Scholar] [CrossRef] [PubMed]
  102. Yamasaki, H.; Aonuma, K.; Shinoda, Y.; Komatsu, Y.; Masuda, K.; Hashimoto, N.; Sai, E.; Yamagami, F.; Okabe, Y.; Tsumagari, Y.; et al. Initial Result of Antrum Pulmonary Vein Isolation Using the Radiofrequency Hot-Balloon Catheter With Single-Shot Technique. JACC Clin. Electrophysiol. 2019, 5, 354–363. [Google Scholar] [CrossRef] [PubMed]
  103. Nakahara, S.; Wakamatsu, Y.; Fukuda, R.; Hori, Y.; Nishiyama, N.; Sato, H.; Nagashima, K.; Mizutani, Y.; Ishikawa, T.; Kobayashi, S.; et al. Utility of hot-balloon-based pulmonary vein isolation under balloon surface temperature monitoring: First clinical experience. J. Cardiovasc. Electrophysiol. 2021, 32, 2625–2635. [Google Scholar] [CrossRef] [PubMed]
  104. Sohara, H.; Satake, S.; Takeda, H.; Yamaguchi, Y.; Nagasu, N. Prevalence of Esophageal Ulceration After Atrial Fibrillation Ablation with the Hot Balloon Ablation Catheter: What is the Value of Esophageal Cooling? J. Cardiovasc. Electrophysiol. 2014, 25, 686–692. [Google Scholar] [CrossRef]
  105. Peng, X.; Liu, X.; Tian, H.; Chen, Y.; Li, X. Effects of Hot Balloon vs. Cryoballoon Ablation for Atrial Fibrillation: A Systematic Review, Meta-Analysis, and Meta-Regression. Front. Cardiovasc. Med. 2021, 8, 787270. [Google Scholar] [CrossRef]
  106. Hojo, R.; Fukamizu, S.; Tokioka, S.; Inagaki, D.; Miyazawa, S.; Kawamura, I.; Kitamura, T.; Sakurada, H.; Hiraoka, M. Comparison of touch-up ablation rate and pulmonary vein isolation durability between hot balloon and cryoballoon. J. Cardiovasc. Electrophysiol. 2020, 31, 1298–1306. [Google Scholar] [CrossRef]
  107. Wu, G.; Huang, H.; Cai, L.; Yang, Y.; Liu, X.; Yu, B.; Tang, Y.; Jiang, H.; Huang, C.; Investigators, C.S. Long-term observation of catheter ablation vs. pharmacotherapy in the management of persistent and long-standing persistent atrial fibrillation (CAPA study). Europace 2021, 23, 731–739. [Google Scholar] [CrossRef]
  108. Yang, G.; Zheng, L.; Jiang, C.; Fan, J.; Liu, X.; Zhan, X.; Li, J.; Wang, L.; Yang, H.; Zhu, W.; et al. Circumferential Pulmonary Vein Isolation Plus Low-Voltage Area Modification in Persistent Atrial Fibrillation: The STABLE-SR-II Trial. JACC Clin. Electrophysiol. 2022, 8, 882–891. [Google Scholar] [CrossRef]
  109. Junarta, J.; Siddiqui, M.U.; Riley, J.M.; Dikdan, S.J.; Patel, A.; Frisch, D.R. Low-voltage area substrate modification for atrial fibrillation ablation: A systematic review and meta-analysis of clinical trials. Europace 2022, euac089. [Google Scholar] [CrossRef]
  110. Hwang, J.; Park, H.-S.; Han, S.; Lee, C.H.; Kim, I.-C.; Cho, Y.-K.; Yoon, H.-J.; Chung, J.W.; Kim, H.; Nam, C.-W.; et al. Ablation of persistent atrial fibrillation based on high density voltage mapping and complex fractionated atrial electrograms: A randomized controlled trial. Medicine 2021, 100, e26702. [Google Scholar] [CrossRef]
  111. Martin, C.A.; Curtain, J.P.; Gajendragadkar, P.R.; Begley, D.A.; Fynn, S.P.; Grace, A.A.; Heck, P.M.; Virdee, M.S.; Agarwal, S. Ablation of Complex Fractionated Electrograms Improves Outcome in Persistent Atrial Fibrillation of Over 2 Years’ Duration. J. Atr. Fibrillation 2018, 10, 1607. [Google Scholar] [CrossRef]
  112. Fink, T.; Schlüter, M.; Heeger, C.-H.; Lemes, C.; Maurer, T.; Reissmann, B.; Riedl, J.; Rottner, L.; Santoro, F.; Schmidt, B.; et al. Stand-Alone Pulmonary Vein Isolation Versus Pulmonary Vein Isolation With Additional Substrate Modification as Index Ablation Procedures in Patients With Persistent and Long-Standing Persistent Atrial Fibrillation: The Randomized Alster-Lost-AF Trial (Ablation at St. Georg Hospital for Long-Standing Persistent Atrial Fibrillation). Circ. Arrhythmia Electrophysiol. 2017, 10, e005114. [Google Scholar] [CrossRef]
  113. Kim, T.; Uhm, J.; Kim, J.-Y.; Joung, B.; Lee, M.-H.; Pak, H. Does Additional Electrogram-Guided Ablation After Linear Ablation Reduce Recurrence After Catheter Ablation for Longstanding Persistent Atrial Fibrillation? A Prospective Randomized Study. J. Am. Heart Assoc. 2017, 6, e004811. [Google Scholar] [CrossRef]
  114. Kochhäuser, S.; Verma, A.; Dalvi, R.; Suszko, A.; Alipour, P.; Sanders, P.; Champagne, J.; Macle, L.; Nair, G.M.; Calkins, H.; et al. Spatial Relationships of Complex Fractionated Atrial Electrograms and Continuous Electrical Activity to Focal Electrical Sources: Implications for Substrate Ablation in Human Atrial Fibrillation. JACC Clin. Electrophysiol. 2017, 3, 1220–1228. [Google Scholar] [CrossRef]
  115. Baher, A.; Buck, B.; Fanarjian, M.; Mounsey, J.P.; Gehi, A.; Chung, E.; Akar, F.G.; Webber, C.L., Jr.; Akar, J.G.; Hummel, J.P. Recurrence quantification analysis of complex-fractionated electrograms differentiates active and passive sites during atrial fibrillation. J. Cardiovasc. Electrophysiol. 2019, 30, 2229–2238. [Google Scholar] [CrossRef]
  116. Lee, J.M.; Shim, J.; Park, J.; Yu, H.T.; Kim, T.-H.; Park, J.-K.; Uhm, J.-S.; Kim, J.-B.; Joung, B.; Lee, M.-H.; et al. The Electrical Isolation of the Left Atrial Posterior Wall in Catheter Ablation of Persistent Atrial Fibrillation. JACC Clin. Electrophysiol. 2019, 5, 1253–1261. [Google Scholar] [CrossRef]
  117. Kanitsoraphan, C.; Rattanawong, P.; Techorueangwiwat, C.; Kewcharoen, J.; Mekritthikrai, R.; Prasitlumkum, N.; Shah, P.; El Masry, H. The efficacy of posterior wall isolation in atrial fibrillation ablation: A systematic review and meta-analysis of randomized controlled trials. J. Arrhythmia 2022, 38, 275–286. [Google Scholar] [CrossRef]
  118. Valderrábano, M.; Peterson, L.E.; Swarup, V.; Schurmann, P.A.; Makkar, A.; Doshi, R.N.; DeLurgio, D.; Athill, C.A.; Ellenbogen, K.A.; Natale, A.; et al. Effect of Catheter Ablation With Vein of Marshall Ethanol Infusion vs Catheter Ablation Alone on Persistent Atrial Fibrillation: The VENUS Randomized Clinical Trial. JAMA 2020, 324, 1620–1628. [Google Scholar] [CrossRef]
  119. Haldar, S.; Khan, H.R.; Boyalla, V.; Kralj-Hans, I.; Jones, S.; Lord, J.; Onyimadu, O.; Satishkumar, A.; Bahrami, T.; De Souza, A.; et al. Catheter ablation vs. thoracoscopic surgical ablation in long-standing persistent atrial fibrillation: CASA-AF randomized controlled trial. Eur. Heart J. 2020, 41, 4471–4480. [Google Scholar] [CrossRef]
  120. Marrouche, N.F.; Wazni, O.; McGann, C.; Greene, T.; Dean, J.M.; Dagher, L.; Kholmovski, E.; Mansour, M.; Marchlinski, F.; Wilber, D.; et al. Effect of MRI-Guided Fibrosis Ablation vs Conventional Catheter Ablation on Atrial Arrhythmia Recurrence in Patients With Persistent Atrial Fibrillation: The DECAAF II Randomized Clinical Trial. JAMA 2022, 327, 2296–2305. [Google Scholar] [CrossRef]
  121. Tilz, R.R.; Yalin, K.; Lyan, E.; Heeger, C.; Schlüter, M.; Fink, T.; Sciacca, V.; Liosis, S.; Kuck, K.; Popescu, S.; et al. Stand-alone Focal Impulse and Rotor Modulation (FIRM) ablation versus second-generation cryoballoon pulmonary vein isolation for paroxysmal atrial fibrillation. J. Cardiovasc. Electrophysiol. 2022, 33, 1678–1686. [Google Scholar] [CrossRef] [PubMed]
  122. Tilz, R.R.; Lenz, C.; Sommer, P.; Roza, M.-S.; Sarver, A.E.; Williams, C.G.; Heeger, C.; Hindricks, G.; Vogler, J.; Eitel, C. Focal Impulse and Rotor Modulation Ablation vs. Pulmonary Vein isolation for the treatment of paroxysmal Atrial Fibrillation: Results from the FIRMAP AF study. Europace 2021, 23, 722–730. [Google Scholar] [CrossRef] [PubMed]
  123. Spitzer, S.G.; Károlyi, L.; Rämmler, C.; Scharfe, F.; Weinmann, T.; Zieschank, M.; Langbein, A. Treatment of Recurrent Nonparoxysmal Atrial Fibrillation Using Focal Impulse and Rotor Mapping (FIRM)-Guided Rotor Ablation: Early Recurrence and Long-Term Outcomes. J. Cardiovasc. Electrophysiol. 2017, 28, 31–38. [Google Scholar] [CrossRef] [PubMed]
  124. Hsieh, Y.; Lin, Y.; Lo, M.; Chen, Y.; Lin, C.; Lin, C.; Chung, F.; Lo, L.; Chang, S.; Chao, T.; et al. Optimal substrate modification strategies using catheter ablation in patients with persistent atrial fibrillation: 3-year follow-up outcomes. J. Cardiovasc. Electrophysiol. 2021, 32, 1561–1571. [Google Scholar] [CrossRef]
  125. Romero, J.; Gabr, M.; Alviz, I.; Briceno, D.; Diaz, J.C.; Rodriguez, D.; Patel, K.; Polanco, D.; Trivedi, C.; Mohanty, S.; et al. Focal impulse and rotor modulation guided ablation versus pulmonary vein isolation for atrial fibrillation: A meta-analysis of head-to-head comparative studies. J. Cardiovasc. Electrophysiol. 2021, 32, 1822–1832. [Google Scholar] [CrossRef]
  126. Atienza, F.; Almendral, J.; Ormaetxe, J.M.; Moya, A.; Martínez-Alday, J.D.; Hernández-Madrid, A.; Castellanos, E.; Arribas, F.; Arias, M.; Tercedor, L.; et al. Comparison of Radiofrequency Catheter Ablation of Drivers and Circumferential Pulmonary Vein Isolation in Atrial Fibrillation: A noninferiority randomized multicenter RADAR-AF trial. J. Am. Coll. Cardiol. 2014, 64, 2455–2467. [Google Scholar] [CrossRef]
  127. Kumagai, K.; Minami, K.; Sugai, Y.; Oshima, S. Evaluation of the atrial substrate based on low-voltage areas and dominant frequencies after pulmonary vein isolation in nonparoxysmal atrial fibrillation. J. Arrhythmia 2018, 34, 230–238. [Google Scholar] [CrossRef]
  128. Kumagai, K.; Minami, K.; Sugai, Y.; Sumiyoshi, T.; Komaru, T. Effect of ablation at high-dominant frequency sites overlapping with low-voltage areas after pulmonary vein isolation of nonparoxysmal atrial fibrillation. J. Cardiovasc. Electrophysiol. 2019, 30, 1850–1859. [Google Scholar] [CrossRef]
  129. Li, X.; Chu, G.S.; Almeida, T.P.; Vanheusden, F.J.; Salinet, J.; Dastagir, N.; Mistry, A.R.; Vali, Z.; Sidhu, B.; Stafford, P.J.; et al. Automatic Extraction of Recurrent Patterns of High Dominant Frequency Mapping During Human Persistent Atrial Fibrillation. Front. Physiol. 2021, 12, 649486. [Google Scholar] [CrossRef]
  130. Wu, S.; Li, H.; Yi, S.; Yao, J.; Chen, X. Comparing the efficacy of catheter ablation strategies for persistent atrial fibrillation: A Bayesian analysis of randomized controlled trials. J. Interv. Card. Electrophysiol. 2022, 1–14. [Google Scholar] [CrossRef]
  131. Hao, J.; Xi, Y.; Chen, W.; Liang, Y.; Lin, Z.; Wei, W. Hybrid ablation procedure for the treatment of nonparoxysmal atrial fibrillation: A systematic review and meta-analysis. Pacing Clin. Electrophysiol. 2022. [Google Scholar] [CrossRef]
  132. Delurgio, D.B.; Crossen, K.J.; Gill, J.; Blauth, C.; Oza, S.R.; Magnano, A.R.; Mostovych, M.A.; Halkos, M.E.; Tschopp, D.R.; Kerendi, F.; et al. Hybrid Convergent Procedure for the Treatment of Persistent and Long-Standing Persistent Atrial Fibrillation: Results of CONVERGE Clinical Trial. Circ. Arrhythmia Electrophysiol. 2020, 13, e009288. [Google Scholar] [CrossRef]
  133. Kodali, S.; Santangeli, P. How, When, and Why: High-Density Mapping of Atrial Fibrillation. Card. Electrophysiol. Clin. 2020, 12, 155–165. [Google Scholar] [CrossRef]
  134. Marrouche, N.F.; Brachmann, J.; Andresen, D.; Siebels, J.; Boersma, L.; Jordaens, L.; Merkely, B.; Pokushalov, E.; Sanders, P.; Proff, J.; et al. Catheter Ablation for Atrial Fibrillation with Heart Failure. N. Engl. J. Med. 2018, 378, 417–427. [Google Scholar] [CrossRef]
  135. Sohns, C.; Zintl, K.; Zhao, Y.; Dagher, L.; Andresen, D.; Siebels, J.; Wegscheider, K.; Sehner, S.; Boersma, L.; Merkely, B.; et al. Impact of Left Ventricular Function and Heart Failure Symptoms on Outcomes Post Ablation of Atrial Fibrillation in Heart Failure: CASTLE-AF Trial. Circ. Arrhythmia Electrophysiol. 2020, 13, e008461. [Google Scholar] [CrossRef]
  136. Brachmann, J.; Sohns, C.; Andresen, D.; Siebels, J.; Sehner, S.; Boersma, L.; Merkely, B.; Pokushalov, E.; Sanders, P.; Schunkert, H.; et al. Atrial Fibrillation Burden and Clinical Outcomes in Heart Failure: The CASTLE-AF Trial. JACC Clin. Electrophysiol. 2021, 7, 594–603. [Google Scholar] [CrossRef]
  137. Noseworthy, P.A.; Van Houten, H.K.; Gersh, B.J.; Packer, D.L.; Friedman, P.A.; Shah, N.D.; Dunlay, S.M.; Siontis, K.C.; Piccini, J.P.; Yao, X. Generalizability of the CASTLE-AF trial: Catheter ablation for patients with atrial fibrillation and heart failure in routine practice. Heart Rhythm 2020, 17, 1057–1065. [Google Scholar] [CrossRef]
  138. Parkash, R.; Wells, G.A.; Rouleau, J.; Talajic, M.; Essebag, V.; Skanes, A.; Wilton, S.B.; Verma, A.; Healey, J.S.; Sterns, L.; et al. Randomized Ablation-Based Rhythm-Control Versus Rate-Control Trial in Patients With Heart Failure and Atrial Fibrillation: Results from the RAFT-AF trial. Circulation 2022, 145, 1693–1704. [Google Scholar] [CrossRef]
  139. Prabhu, S.; Taylor, A.J.; Costello, B.T.; Kaye, D.M.; McLellan, A.J.; Voskoboinik, A.; Sugumar, H.; Lockwood, S.M.; Stokes, M.B.; Pathik, B.; et al. Catheter Ablation Versus Medical Rate Control in Atrial Fibrillation and Systolic Dysfunction: The CAMERA-MRI Study. J. Am. Coll. Cardiol. 2017, 70, 1949–1961. [Google Scholar] [CrossRef]
  140. Sugumar, H.; Prabhu, S.; Costello, B.; Chieng, D.; Azzopardi, S.; Voskoboinik, A.; Parameswaran, R.; Wong, G.R.; Anderson, R.; Al-Kaisey, A.M.; et al. Catheter Ablation Versus Medication in Atrial Fibrillation and Systolic Dysfunction: Late Outcomes of CAMERA-MRI Study. JACC Clin. Electrophysiol. 2020, 6, 1721–1731. [Google Scholar] [CrossRef]
  141. Kuck, K.-H.; Merkely, B.; Zahn, R.; Arentz, T.; Seidl, K.; Schlüter, M.; Tilz, R.R.; Piorkowski, C.; Gellér, L.; Kleemann, T.; et al. Catheter Ablation Versus Best Medical Therapy in Patients With Persistent Atrial Fibrillation and Congestive Heart Failure: The Randomized AMICA Trial. Circ. Arrhythmia Electrophysiol. 2019, 12, e007731. [Google Scholar] [CrossRef]
  142. Gu, G.; Wu, J.; Gao, X.; Liu, M.; Jin, C.; Xu, Y. Catheter ablation of atrial fibrillation in patients with heart failure and preserved ejection fraction: A meta-analysis. Clin. Cardiol. 2022, 45, 786–793. [Google Scholar] [CrossRef]
  143. Androulakis, E.; Sohrabi, C.; Briasoulis, A.; Bakogiannis, C.; Saberwal, B.; Siasos, G.; Tousoulis, D.; Ahsan, S.; Papageorgiou, N. Catheter Ablation for Atrial Fibrillation in Patients with Heart Failure with Preserved Ejection Fraction: A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 288. [Google Scholar] [CrossRef]
  144. Zhang, H.; Yu, M.; Xia, Y.; Li, X.; Liu, J.; Fang, P. The differences of atrial thrombus locations and variable response to anticoagulation in nonvalvular atrial fibrillation with ventricular cardiomyopathy. J. Arrhythmia 2020, 36, 1016–1022. [Google Scholar] [CrossRef]
  145. Al-Saady, N.M.; Obel, O.A.; Camm, A.J. Left atrial appendage: Structure, function, and role in thromboembolism. Heart 1999, 82, 547–554. [Google Scholar] [CrossRef]
  146. Whitlock, R.P.; Belley-Cote, E.P.; Paparella, D.; Healey, J.S.; Brady, K.; Sharma, M.; Reents, W.; Budera, P.; Baddour, A.J.; Fila, P.; et al. Left Atrial Appendage Occlusion during Cardiac Surgery to Prevent Stroke. N. Engl. J. Med. 2021, 384, 2081–2091. [Google Scholar] [CrossRef]
  147. Nso, N.; Nassar, M.; Zirkiyeva, M.; Lakhdar, S.; Shaukat, T.; Guzman, L.; Alshamam, M.; Foster, A.; Bhangal, R.; Badejoko, S.; et al. Outcomes of cardiac surgery with left atrial appendage occlusion versus no Occlusion, direct oral Anticoagulants, and vitamin K Antagonists: A systematic review with Meta-analysis. IJC Heart Vasc. 2022, 40, 100998. [Google Scholar] [CrossRef]
  148. Radinovic, A.; Falasconi, G.; Marzi, A.; D’Angelo, G.; Limite, L.; Paglino, G.; Peretto, G.; Frontera, A.; Fierro, N.; Sala, S.; et al. Long-term outcome of left atrial appendage occlusion with multiple devices. Int. J. Cardiol. 2021, 344, 66–72. [Google Scholar] [CrossRef]
  149. Osmancik, P.; Herman, D.; Neuzil, P.; Hala, P.; Taborsky, M.; Kala, P.; Poloczek, M.; Stasek, J.; Haman, L.; Branny, M.; et al. Left Atrial Appendage Closure Versus Direct Oral Anticoagulants in High-Risk Patients With Atrial Fibrillation. J. Am. Coll. Cardiol. 2020, 75, 3122–3135. [Google Scholar] [CrossRef] [PubMed]
  150. Bayard, Y.L.; Omran, H.; Neuzil, P.; Thuesen, L.; Pichler, M.; Rowland, E.; Ramondo, A.; Ruzyllo, W.; Budts, W.; Montalescot, G.; et al. PLAATO (Percutaneous Left Atrial Appendage Transcatheter Occlusion) for prevention of cardioembolic stroke in non-anticoagulation eligible atrial fibrillation patients: Results from the European PLAATO study. EuroIntervention 2010, 6, 220–226. [Google Scholar] [CrossRef] [PubMed]
  151. Reddy, V.Y.; Sievert, H.; Halperin, J.; Doshi, S.K.; Buchbinder, M.; Neuzil, P.; Huber, K.; Whisenant, B.; Kar, S.; Swarup, V.; et al. Percutaneous Left Atrial Appendage Closure vs Warfarin for Atrial Fibrillation: A randomized clinical trial. JAMA 2014, 312, 1988–1998. [Google Scholar] [CrossRef] [PubMed]
  152. Holmes, D.R., Jr.; Reddy, V.Y.; Gordon, N.T.; Delurgio, D.; Doshi, S.K.; Desai, A.J.; Stone, J.E., Jr.; Kar, S. Long-Term Safety and Efficacy in Continued Access Left Atrial Appendage Closure Registries. J. Am. Coll. Cardiol. 2019, 74, 2878–2889. [Google Scholar] [CrossRef]
  153. Price, M.J.; Slotwiner, D.; Du, C.; Freeman, J.V.; Turi, Z.; Rammohan, C.; Kusumoto, F.M.; Kavinsky, C.; Akar, J.; Varosy, P.D.; et al. Clinical Outcomes at 1 Year Following Transcatheter Left Atrial Appendage Occlusion in the United States. JACC Cardiovasc. Interv. 2022, 15, 741–750. [Google Scholar] [CrossRef]
  154. Alli, O.; Doshi, S.; Kar, S.; Reddy, V.; Sievert, H.; Mullin, C.; Swarup, V.; Whisenant, B.; Holmes, D., Jr. Quality of Life Assessment in the Randomized PROTECT AF (Percutaneous Closure of the Left Atrial Appendage Versus Warfarin Therapy for Prevention of Stroke in Patients With Atrial Fibrillation) Trial of Patients at Risk for Stroke With Nonvalvular Atrial Fibrillation. J. Am. Coll. Cardiol. 2013, 61, 1790–1798. [Google Scholar] [CrossRef]
  155. Coisne, A.; Pilato, R.; Brigadeau, F.; Klug, D.; Marquie, C.; Souissi, Z.; Richardson, M.; Mouton, S.; Polge, A.-S.; Lancellotti, P.; et al. Percutaneous left atrial appendage closure improves left atrial mechanical function through Frank–Starling mechanism. Heart Rhythm 2017, 14, 710–716. [Google Scholar] [CrossRef]
  156. Viles-Gonzalez, J.F.; Kar, S.; Douglas, P.; Dukkipati, S.; Feldman, T.; Horton, R.; Holmes, D.; Reddy, V.Y. The Clinical Impact of Incomplete Left Atrial Appendage Closure With the Watchman Device in Patients With Atrial Fibrillation: A PROTECT AF (Percutaneous Closure of the Left Atrial Appendage Versus Warfarin Therapy for Prevention of Stroke in Patients With Atrial Fibrillation) Substudy. J. Am. Coll. Cardiol. 2012, 59, 923–929. [Google Scholar] [CrossRef]
  157. Dukkipati, S.R.; Holmes, D.R., Jr.; Doshi, S.K.; Kar, S.; Singh, S.M.; Gibson, D.; Price, M.J.; Natale, A.; Mansour, M.; Sievert, H.; et al. Impact of Peridevice Leak on 5-Year Outcomes After Left Atrial Appendage Closure. J. Am. Coll. Cardiol. 2022, 80, 469–483. [Google Scholar] [CrossRef]
  158. Dukkipati, S.R.; Kar, S.; Holmes, D.R.; Doshi, S.K.; Swarup, V.; Gibson, D.N.; Maini, B.; Gordon, N.T.; Main, M.L.; Reddy, V.Y. Device-Related Thrombus After Left Atrial Appendage Closure: Incidence, Predictors, and Outcomes. Circulation 2018, 138, 874–885. [Google Scholar] [CrossRef]
  159. Simard, T.; Jung, R.G.; Lehenbauer, K.; Piayda, K.; Pracoń, R.; Jackson, G.G.; Flores-Umanzor, E.; Faroux, L.; Korsholm, K.; Chun, J.K.; et al. Predictors of Device-Related Thrombus Following Percutaneous Left Atrial Appendage Occlusion. J. Am. Coll. Cardiol. 2021, 78, 297–313. [Google Scholar] [CrossRef]
  160. Murtaza, G.; Turagam, M.K.; Dar, T.; Akella, K.; Yarlagadda, B.; Gloekler, S.; Meier, B.; Saw, J.; Kim, J.S.; Lim, H.E.; et al. Left Atrial Appendage Occlusion Device Embolization (The LAAODE Study): Understanding the Timing and Clinical Consequences from a Worldwide Experience. J. Atr. Fibrillation 2021, 13, 2516. [Google Scholar] [CrossRef]
  161. Cruz-González, I.; Korsholm, K.; Trejo-Velasco, B.; Thambo, J.B.; Mazzone, P.; Rioufol, G.; Grygier, M.; Möbius-Winkler, S.; Betts, T.; Meincke, F.; et al. Procedural and Short-Term Results With the New Watchman FLX Left Atrial Appendage Occlusion Device. JACC: Cardiovasc. Interv. 2020, 13, 2732–2741. [Google Scholar] [CrossRef]
  162. Korsholm, K.; Samaras, A.; Andersen, A.; Jensen, J.M.; Nielsen-Kudsk, J.E. The Watchman FLX Device: First European Experience and Feasibility of Intracardiac Echocardiography to Guide Implantation. JACC Clin. Electrophysiol. 2020, 6, 1633–1642. [Google Scholar] [CrossRef]
  163. Kar, S.; Doshi, S.K.; Sadhu, A.; Horton, R.; Osorio, J.; Ellis, C.; Stone, J., Jr.; Shah, M.; Dukkipati, S.R.; Adler, S.; et al. Primary Outcome Evaluation of a Next-Generation Left Atrial Appendage Closure Device: Results From the PINNACLE FLX Trial. Circulation 2021, 143, 1754–1762. [Google Scholar] [CrossRef]
  164. Ellis, C.R.; Jackson, G.G.; Kanagasundram, A.N.; Mansour, M.; Sutton, B.; Houle, V.M.; Kar, S.; Doshi, S.; Osorio, J. Left atrial appendage closure in patients with prohibitive anatomy: Insights from PINNACLE FLX. Heart Rhythm 2021, 18, 1153–1161. [Google Scholar] [CrossRef]
  165. Paitazoglou, C.; Meincke, F.; Bergmann, M.W.; Eitel, I.; Fink, T.; Vireca, E.; Wohlmuth, P.; Veliqi, E.; Willems, S.; Markiewicz, A.; et al. The ALSTER-FLX Registry: 3-Month outcomes after left atrial appendage occlusion using a next-generation device, a matched-pair analysis to EWOLUTION. Heart Rhythm 2022, 19, 917–926. [Google Scholar] [CrossRef]
  166. Vizzari, G.; Grasso, C.; Sardone, A.; Mazzone, P.; Laterra, G.; Frazzetto, M.; Sacchetta, G.; Micari, A.; Tamburino, C.; Contarini, M. Real-world experience with the new Watchman FLX device: Data from two high-volume Sicilian centers. The FLX-iEST registry. Catheter. Cardiovasc. Interv. 2022, 100, 154–160. [Google Scholar] [CrossRef]
  167. Galea, R.; Mahmoudi, K.; Gräni, C.; Elhadad, S.; Huber, A.T.; Heg, D.; Siontis, G.C.M.; Brugger, N.; Sebag, F.; Windecker, S.; et al. Watchman FLX vs. Watchman 2.5 in a Dual-Center Left Atrial Appendage Closure Cohort: The WATCH-DUAL study. Europace 2022, euac021. [Google Scholar] [CrossRef]
  168. Meier, B.; Palacios, I.; Windecker, S.; Rotter, M.; Cao, Q.-L.; Keane, D.; Ruiz, C.E.; Hijazi, Z.M. Transcatheter left atrial appendage occlusion with Amplatzer devices to obviate anticoagulation in patients with atrial fibrillation. Catheter. Cardiovasc. Interv. 2003, 60, 417–422. [Google Scholar] [CrossRef]
  169. Park, J.-W.; Bethencourt, A.; Sievert, H.; Santoro, G.; Meier, B.; Walsh, K.; Lopez-Minquez, J.R.; Meerkin, D.; Valdés, M.; Ormerod, O.; et al. Left atrial appendage closure with amplatzer cardiac plug in atrial fibrillation: Initial european experience. Catheter. Cardiovasc. Interv. 2011, 77, 700–706. [Google Scholar] [CrossRef]
  170. Lam, Y.-Y.; Yip, G.; Yu, C.-M.; Chan, W.W.; Cheng, B.C.; Yan, B.P.; Clugston, R.; Yong, G.; Gattorna, T.; Paul, V. Left atrial appendage closure with Amplatzer cardiac plug for stroke prevention in atrial fibrillation: Initial Asia-Pacific experience. Catheter. Cardiovasc. Interv. 2012, 79, 794–800. [Google Scholar] [CrossRef]
  171. Urena, M.; Rodés-Cabau, J.; Freixa, X.; Saw, J.; Webb, J.G.; Freeman, M.; Horlick, E.; Osten, M.; Chan, A.; Marquis, J.-F.; et al. Percutaneous Left Atrial Appendage Closure With the AMPLATZER Cardiac Plug Device in Patients With Nonvalvular Atrial Fibrillation and Contraindications to Anticoagulation Therapy. J. Am. Coll. Cardiol. 2013, 62, 96–102. [Google Scholar] [CrossRef] [PubMed]
  172. Tzikas, A.; Shakir, S.; Gafoor, S.; Omran, H.; Berti, S.; Santoro, G.; Kefer, J.; Landmesser, U.; Nielsen-Kudsk, J.E.; Cruz-Gonzalez, I.; et al. Left atrial appendage occlusion for stroke prevention in atrial fibrillation: Multicentre experience with the AMPLATZER Cardiac Plug. EuroIntervention 2016, 11, 1170–1179. [Google Scholar] [CrossRef] [PubMed]
  173. Santoro, G.; Meucci, F.; Stolcova, M.; Rezzaghi, M.; Mori, F.; Palmieri, C.; Paradossi, U.; Pastormerlo, L.E.; Rosso, G.; Berti, S. Percutaneous left atrial appendage occlusion in patients with non-valvular atrial fibrillation: Implantation and up to four years follow-up of the AMPLATZER Cardiac Plug. EuroIntervention 2016, 11, 1188–1194. [Google Scholar] [CrossRef] [PubMed]
  174. Freixa, X.; Gafoor, S.; Regueiro, A.; Cruz-Gonzalez, I.; Shakir, S.; Omran, H.; Berti, S.; Santoro, G.; Kefer, J.; Landmesser, U.; et al. Comparison of Efficacy and Safety of Left Atrial Appendage Occlusion in Patients Aged <75 to ≥75 Years. Am. J. Cardiol. 2016, 117, 84–90. [Google Scholar] [CrossRef]
  175. Kefer, J.; Tzikas, A.; Freixa, X.; Shakir, S.; Gafoor, S.; Nielsen-Kudsk, J.E.; Berti, S.; Santoro, G.; Aminian, A.; Landmesser, U.; et al. Impact of chronic kidney disease on left atrial appendage occlusion for stroke prevention in patients with atrial fibrillation. Int. J. Cardiol. 2016, 207, 335–340. [Google Scholar] [CrossRef]
  176. Plicht, B.; Konorza, T.F.; Kahlert, P.; Al-Rashid, F.; Kaelsch, H.; Jánosi, R.A.; Buck, T.; Bachmann, H.S.; Siffert, W.; Heusch, G.; et al. Risk Factors for Thrombus Formation on the Amplatzer Cardiac Plug After Left Atrial Appendage Occlusion. JACC: Cardiovasc. Interv. 2013, 6, 606–613. [Google Scholar] [CrossRef]
  177. Clemente, A.; Avogliero, F.; Berti, S.; Paradossi, U.; Jamagidze, G.; Rezzaghi, M.; Della Latta, D.; Chiappino, D. Multimodality imaging in preoperative assessment of left atrial appendage transcatheter occlusion with the Amplatzer Cardiac Plug. Eur. Heart J. Cardiovasc. Imaging 2015, 16, 1276–1287. [Google Scholar] [CrossRef]
  178. Saw, J.; Tzikas, A.; Shakir, S.; Gafoor, S.; Omran, H.; Nielsen-Kudsk, J.E.; Kefer, J.; Aminian, A.; Berti, S.; Santoro, G.; et al. Incidence and Clinical Impact of Device-Associated Thrombus and Peri-Device Leak Following Left Atrial Appendage Closure With the Amplatzer Cardiac Plug. JACC Cardiovasc. Interv. 2017, 10, 391–399. [Google Scholar] [CrossRef]
  179. Hildick-Smith, D.; Landmesser, U.; Camm, A.J.; Diener, H.-C.; Paul, V.; Schmidt, B.; Settergren, M.; Teiger, E.; Nielsen-Kudsk, J.E.; Tondo, C. Left atrial appendage occlusion with the Amplatzer™ Amulet™ device: Full results of the prospective global observational study. Eur. Heart J. 2020, 41, 2894–2901. [Google Scholar] [CrossRef]
  180. Tarantini, G.; D’Amico, G.; Schmidt, B.; Mazzone, P.; Berti, S.; Fischer, S.; Lund, J.; Montorfano, M.; Della Bella, P.; Lam, S.C.C.; et al. The Impact of CHA2DS2-VASc and HAS-BLED Scores on Clinical Outcomes in the Amplatzer Amulet Study. JACC Cardiovasc. Interv. 2020, 13, 2099–2108. [Google Scholar] [CrossRef]
  181. Freixa, X.; Schmidt, B.; Mazzone, P.; Berti, S.; Fischer, S.; Lund, J.; Montorfano, M.; Della Bella, P.; Lam, S.C.C.; Cruz-Gonzalez, I.; et al. Comparative data on left atrial appendage occlusion efficacy and clinical outcomes by age group in the Amplatzer™ Amulet™ Occluder Observational Study. Europace 2021, 23, 238–246. [Google Scholar] [CrossRef]
  182. Aminian, A.; Schmidt, B.; Mazzone, P.; Berti, S.; Fischer, S.; Montorfano, M.; Lam, S.C.C.; Lund, J.; Asch, F.M.; Gage, R.; et al. Incidence, Characterization, and Clinical Impact of Device-Related Thrombus Following Left Atrial Appendage Occlusion in the Prospective Global AMPLATZER Amulet Observational Study. JACC Cardiovasc. Interv. 2019, 12, 1003–1014. [Google Scholar] [CrossRef]
  183. Kleinecke, C.; Cheikh-Ibrahim, M.; Schnupp, S.; Fankhauser, M.; Nietlispach, F.; Park, J.; Brachmann, J.; Windecker, S.; Meier, B.; Gloekler, S. Long-term clinical outcomes of Amplatzer cardiac plug versus Amulet occluders for left atrial appendage closure. Catheter. Cardiovasc. Interv. 2020, 96, E324–E331. [Google Scholar] [CrossRef]
  184. Gloekler, S.; Fürholz, M.; de Marchi, S.; Kleinecke, C.; Streit, S.R.; Buffle, E.; Fankhauser, M.; Häner, J.D.; Nietlispach, F.; Galea, R.; et al. Left atrial appendage closure versus medical therapy in patients with atrial fibrillation: The APPLY study. EuroIntervention 2020, 16, e767–e774. [Google Scholar] [CrossRef]
  185. Nielsen-Kudsk, J.E.; Korsholm, K.; Damgaard, D.; Valentin, J.B.; Diener, H.-C.; Camm, A.J.; Johnsen, S.P. Clinical Outcomes Associated With Left Atrial Appendage Occlusion Versus Direct Oral Anticoagulation in Atrial Fibrillation. JACC Cardiovasc. Interv. 2021, 14, 69–78. [Google Scholar] [CrossRef]
  186. Ledwoch, J.; Franke, J.; Akin, I.; Geist, V.; Weiß, C.; Zeymer, U.; Pleger, S.; Hochadel, M.; Mudra, H.; Senges, J.; et al. WATCHMAN versus ACP or Amulet devices for left atrial appendage occlusion: A sub-analysis of the multicentre LAARGE registry. EuroIntervention 2020, 16, e942–e949. [Google Scholar] [CrossRef]
  187. Kleinecke, C.; Yu, J.; Neef, P.; Buffle, E.; de Marchi, S.; Fuerholz, M.; Nietlispach, F.; Valgimigli, M.; Streit, S.R.; Fankhauser, M.; et al. Clinical outcomes of Watchman vs. Amplatzer occluders for left atrial appendage closure (WATCH at LAAC). Europace 2020, 22, 916–923. [Google Scholar] [CrossRef]
  188. Lakkireddy, D.; Thaler, D.; Ellis, C.R.; Swarup, V.; Sondergaard, L.; Carroll, J.; Gold, M.R.; Hermiller, J.; Diener, H.-C.; Schmidt, B.; et al. Amplatzer Amulet Left Atrial Appendage Occluder Versus Watchman Device for Stroke Prophylaxis (Amulet IDE): A Randomized, Controlled Trial. Circulation 2021, 144, 1543–1552. [Google Scholar] [CrossRef]
  189. Price, M.J.; Gibson, D.N.; Yakubov, S.J.; Schultz, J.C.; Di Biase, L.; Natale, A.; Burkhardt, J.D.; Pershad, A.; Byrne, T.J.; Gidney, B.; et al. Early Safety and Efficacy of Percutaneous Left Atrial Appendage Suture Ligation. J. Am. Coll. Cardiol. 2014, 64, 565–572. [Google Scholar] [CrossRef]
  190. Miller, M.A.; Gangireddy, S.R.; Doshi, S.K.; Aryana, A.; Koruth, J.S.; Sennhauser, S.; D’Avila, A.; Dukkipati, S.R.; Neuzil, P.; Reddy, V.Y. Multicenter study on acute and long-term safety and efficacy of percutaneous left atrial appendage closure using an epicardial suture snaring device. Heart Rhythm 2014, 11, 1853–1859. [Google Scholar] [CrossRef]
  191. Gunda, S.; Reddy, M.; Nath, J.; Nagaraj, H.; Atoui, M.; Rasekh, A.; Ellis, C.R.; Badhwar, N.; Lee, R.J.; Di Biase, L.; et al. Impact of Periprocedural Colchicine on Postprocedural Management in Patients Undergoing a Left Atrial Appendage Ligation Using LARIAT. J. Cardiovasc. Electrophysiol. 2016, 27, 60–64. [Google Scholar] [CrossRef] [PubMed]
  192. Lakkireddy, D.; Afzal, M.R.; Lee, R.J.; Nagaraj, H.; Tschopp, D.; Gidney, B.; Ellis, C.; Altman, E.; Lee, B.; Kar, S.; et al. Short and long-term outcomes of percutaneous left atrial appendage suture ligation: Results from a US multicenter evaluation. Heart Rhythm 2016, 13, 1030–1036. [Google Scholar] [CrossRef] [PubMed]
  193. Afzal, M.R.; Kanmanthareddy, A.; Earnest, M.; Reddy, M.; Atkins, D.; Bommana, S.; Bartus, K.; Rasekh, A.; Han, F.; Badhwar, N.; et al. Impact of left atrial appendage exclusion using an epicardial ligation system (LARIAT) on atrial fibrillation burden in patients with cardiac implantable electronic devices. Heart Rhythm 2015, 12, 52–59. [Google Scholar] [CrossRef] [PubMed]
  194. Turagam, M.; Atkins, D.; Earnest, M.; Lee, R.; Nath, J.; Ferrell, R.; Bartus, K.; Badhwar, N.; Rasekh, A.; Cheng, J.; et al. Anatomical and electrical remodeling with incomplete left atrial appendage ligation: Results from the LAALA-AF registry. J. Cardiovasc. Electrophysiol. 2017, 28, 1433–1442. [Google Scholar] [CrossRef] [PubMed]
  195. Dar, T.; Afzal, M.R.; Yarlagadda, B.; Kutty, S.; Shang, Q.; Gunda, S.; Samanta, A.; Thummaluru, J.; Arukala, K.S.; Kanmanthareddy, A.; et al. Mechanical function of the left atrium is improved with epicardial ligation of the left atrial appendage: Insights from the LAFIT-LARIAT Registry. Heart Rhythm 2018, 15, 955–959. [Google Scholar] [CrossRef] [PubMed]
  196. Kreidieh, B.; Rojas, F.; Schurmann, P.; Dave, A.S.; Kashani, A.; Rodríguez-Mañero, M.; Valderrábano, M. Left Atrial Appendage Remodeling After Lariat Left Atrial Appendage Ligation. Circ. Arrhythmia Electrophysiol. 2015, 8, 1351–1358. [Google Scholar] [CrossRef] [PubMed]
  197. Litwinowicz, R.; Bartus, M.; Burysz, M.; Brzeziński, M.; Suwalski, P.; Kapelak, B.; Vuddanda, V.; Lakkireddy, D.; Lee, R.J.; Trabka, R.; et al. Long term outcomes after left atrial appendage closure with the LARIAT device—Stroke risk reduction over five years follow-up. PLoS ONE 2018, 13, e0208710. [Google Scholar] [CrossRef]
  198. Parikh, V.; Bartus, K.; Litwinowicz, R.; Turagam, M.K.; Sadowski, J.; Kapelak, B.; Bartus, M.; Podolec, J.; Brzezinski, M.; Musat, D.; et al. Long-term clinical outcomes from real-world experience of left atrial appendage exclusion with LARIAT device. J. Cardiovasc. Electrophysiol. 2019, 30, 2849–2857. [Google Scholar] [CrossRef]
  199. Litwinowicz, R.; Bartus, M.; Kapelak, B.; Suwalski, P.; Lakkireddy, D.; Lee, R.J.; Bartus, K. Reduction in risk of stroke and bleeding after left atrial appendage closure with LARIAT device in patients with increased risk of stroke and bleeding: Long term results. Catheter. Cardiovasc. Interv. 2019, 94, 837–842. [Google Scholar] [CrossRef]
  200. Mohanty, S.; Gianni, C.; Trivedi, C.; Gadiyaram, V.; Della Rocca, D.; MacDonald, B.; Horton, R.; Al-Ahmad, A.; Gibson, D.N.; Price, M.; et al. Risk of thromboembolic events after percutaneous left atrial appendage ligation in patients with atrial fibrillation: Long-term results of a multicenter study. Heart Rhythm 2020, 17, 175–181. [Google Scholar] [CrossRef]
  201. Gianni, C.; Di Biase, L.; Trivedi, C.; Mohanty, S.; Gökoğlan, Y.; Güneş, M.F.; Bai, R.; Al-Ahmad, A.; Burkhardt, J.D.; Horton, R.P.; et al. Clinical Implications of Leaks Following Left Atrial Appendage Ligation With the LARIAT Device. JACC Cardiovasc. Interv. 2016, 9, 1051–1057. [Google Scholar] [CrossRef]
  202. Della Rocca, D.G.; Horton, R.P.; Tarantino, N.; Van Niekerk, C.J.; Trivedi, C.; Chen, Q.; Mohanty, S.; Anannab, A.; Murtaza, G.; Akella, K.; et al. Use of a Novel Septal Occluder Device for Left Atrial Appendage Closure in Patients With Postsurgical and Postlariat Leaks or Anatomies Unsuitable for Conventional Percutaneous Occlusion. Circ. Cardiovasc. Interv. 2020, 13, e009227. [Google Scholar] [CrossRef]
  203. Vuddanda, V.L.; Turagam, M.K.; Umale, N.A.; Shah, Z.; Lakkireddy, D.R.; Bartus, K.; McCausland, F.R.; Velagapudi, P.; Mansour, M.; Heist, E.K. Incidence and causes of in-hospital outcomes and 30-day readmissions after percutaneous left atrial appendage closure: A US nationwide retrospective cohort study using claims data. Heart Rhythm 2020, 17, 374–382. [Google Scholar] [CrossRef]
  204. Litwinowicz, R.; Burysz, M.; Mazur, P.; Kapelak, B.; Bartus, M.; Lakkireddy, D.; Lee, R.J.; Malec-Litwinowicz, M.; Bartus, K.; Mazur, P. Endocardial versus epicardial left atrial appendage exclusion for stroke prevention in patients with atrial fibrillation: Midterm follow-up. J. Cardiovasc. Electrophysiol. 2021, 32, 93–101. [Google Scholar] [CrossRef]
  205. Tilz, R.R.; Fink, T.; Bartus, K.; Wong, T.; Vogler, J.; Nentwich, K.; Panniker, S.; Fang, Q.; Piorkowski, C.; Liosis, S.; et al. A collective European experience with left atrial appendage suture ligation using the LARIAT+ device. Europace 2020, 22, 924–931. [Google Scholar] [CrossRef]
  206. Bartus, K.; Gafoor, S.; Tschopp, D.; Foran, J.P.; Tilz, R.; Wong, T.; Lakkireddy, D.; Sievert, H.; Lee, R.J. Left atrial appendage ligation with the next generation LARIAT+ suture delivery device: Early clinical experience. Int. J. Cardiol. 2016, 215, 244–247. [Google Scholar] [CrossRef]
  207. So, C.-Y.; Li, S.; Fu, G.-H.; Chen, W.; Kam, K.K.-H.; Lee, A.P.-W.; Chu, H.-M.; Xu, Y.-W.; Yan, B.P.; Lam, Y.-Y. Procedural and short-term outcomes of occluding large left atrial appendages with the LAmbre device. EuroIntervention 2021, 17, 90–92. [Google Scholar] [CrossRef]
  208. Wang, G.; Kong, B.; Qin, T.; Liu, Y.; Huang, C.; Huang, H. Incidence, risk factors, and clinical impact of peridevice leak following left atrial appendage closure with the LAmbre device—Data from a prospective multicenter clinical study. J. Cardiovasc. Electrophysiol. 2021, 32, 354–359. [Google Scholar] [CrossRef]
  209. Llagostera-Martín, M.; Cubero-Gallego, H.; Mas-Stachurska, A.; Salvatella, N.; Sánchez-Carpintero, A.; Tizon-Marcos, H.; Garcia-Guimaraes, M.; Calvo-Fernandez, A.; Molina, L.; Vaquerizo, B. Left Atrial Appendage Closure with a New Occluder Device: Efficacy, Safety and Mid-Term Performance. J. Clin. Med. 2021, 10, 1421. [Google Scholar] [CrossRef]
  210. Wang, G.; Kong, B.; Liu, Y.; Huang, H. Long-Term Safety and Efficacy of Percutaneous Left Atrial Appendage Closure with the LAmbre Device. J. Interv. Cardiol. 2020, 2020, 6613683. [Google Scholar] [CrossRef]
  211. Schnupp, S.; Liu, X.; Buffle, E.; Gloekler, S.; Mohrez, Y.; Cheikh-Ibrahim, M.; Allakkis, W.; Brachmann, J.; Park, J.; Kleinecke, C. Late clinical outcomes of lambre versus amplatzer occluders for left atrial appendage closure. J. Cardiovasc. Electrophysiol. 2020, 31, 934–942. [Google Scholar] [CrossRef] [PubMed]
  212. Chen, S.; Chun, K.J.; Bordignon, S.; Weise, F.K.; Nagase, T.; Perrotta, L.; Bologna, F.; Schmidt, B. Left atrial appendage occlusion using LAmbre Amulet and Watchman in atrial fibrillation. J. Cardiol. 2019, 73, 299–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Radiofrequency catheter ablation of AF triggers. Although pulmonary vein isolation (yellow circles) remains the cornerstone of AF ablation, the ablation of the left atrial posterior wall (green circles), ganglionated plexuses (red circles), and retrograde ethanol infusion in the vein of Marshall (purple circles) are additional options that may be considered. IVC: inferior vena cava, LIPV: left inferior pulmonary vein, LIGP: left inferior ganglionated plexus, LSPV: left superior pulmonary vein, LSGP: left superior ganglionated plexus, RIGP: right inferior ganglionated plexus, RSGP: right superior ganglionated plexus, RIPV: right inferior pulmonary vein, RSPV: right superior pulmonary vein, SVC: superior vena cava.
Figure 1. Radiofrequency catheter ablation of AF triggers. Although pulmonary vein isolation (yellow circles) remains the cornerstone of AF ablation, the ablation of the left atrial posterior wall (green circles), ganglionated plexuses (red circles), and retrograde ethanol infusion in the vein of Marshall (purple circles) are additional options that may be considered. IVC: inferior vena cava, LIPV: left inferior pulmonary vein, LIGP: left inferior ganglionated plexus, LSPV: left superior pulmonary vein, LSGP: left superior ganglionated plexus, RIGP: right inferior ganglionated plexus, RSGP: right superior ganglionated plexus, RIPV: right inferior pulmonary vein, RSPV: right superior pulmonary vein, SVC: superior vena cava.
Biomedicines 10 02268 g001
Table 1. Landmark randomized clinical trials in patients with atrial fibrillation (AF) and heart failure (HF).
Table 1. Landmark randomized clinical trials in patients with atrial fibrillation (AF) and heart failure (HF).
TrialPatient NumberStudy PopulationControl GroupFollow-upPrimary EndpointOther
CASTLE-AF [134,135,136]363Symptomatic AF
HF (LVEF < 35%)
NYHA class II-IV
ICD
AADs37.8 monthsAll-cause mortality or HF hospitalization
(HR 0.62, 95% CI 0.43–0.87, p = 0.007)
Effect irrespective of LVEF
Less severe functional status led to greater improvement
↓ AF burden
RAFT-AF [138]411High-burden PAF or PeAF
NYHA class II-III
Elevated NT-proBNP
MRC37.4 monthsAll-cause mortality or HF events
(HR 0.71, 95% CI 0.49–1.03, p = 0.066)
↑ LVEF
↓ NT-proBNP
↑ QoL
↑ 6MWT
CAMERA-MRI [139,140]68Persistent AF
LVEF ≤ 45% without identifiable cause
MRC6 monthsChange in cMRI-LVEF
(MD 14.0, 95% CI 8.5–19.5, p < 0.0001)
LVEF normalization
(CA 58% vs. MRC 9%, p = 0.0002)
↓ NYHA class
↓ BNP
↓ LAVi
↓ AF recurrence-burden
AMICA140PeAF
LVEF ≤ 35%
BMT12 monthsIncrease in LVEF
(No difference)
↓ AF recurrence-burden
↔ BNP
↔ 6MWT
↔ QoL
LVEF: left ventricular ejection fraction, NYHA: New York Heart Association, ICD: implantable cardioverter defibrillator, HR: hazard ratio, CI: confidence interval, PAF: paroxysmal AF, PeAF: persistent AF, NT-proBNP: N-terminal pro-hormone brain natriuretic peptide, MRC: medical rate control, QoL: quality of life, 6MWT: 6-min walk test, cMRI: cardiac magnetic resonance imaging, MD: mean difference, CA: catheter ablation, LAVi: left atrial volume index, BMT: best medical therapy. ↓ indicates a decrease, ↑ indicates an increase, ↔ indicates no change
Table 2. Characteristics and clinical outcomes of the most well-studied left atrial appendage (LAA) closure devices.
Table 2. Characteristics and clinical outcomes of the most well-studied left atrial appendage (LAA) closure devices.
ACPAmuletWMWM FLX
DescriptionSelf-expanding, double-disc device consisting of a nitinol mesh with polyester fabricSelf-expanding nitinol 10-strut frame with a 160-μm polyethylene terephthalate fabric mesh cap
Device size, mm16-3016-3421–3320–35
Implant success (%)97.399.195.198.8
Periprocedural MAE (%)5.042.20.5
Device embolization (%)0.80.20.70
DRT (%)4.41.6
(1-year)
3.71.8
Risk factor for DRT Large LAA orifice widthHypercoagulability disorders
Pericardial effusion
Renal impairment
Implantation depth >10 mm from the pulmonary vein limbus
Nonparoxysmal AF
NA
PDL ≥ 3 mm (%)1.91.613.17.4 (>0mm)
All-cause mortality (%)4.2
(1-year)
NA3.6
(5-year)
6.6
(1-year)
Ischemic stroke (%)0.9
(1-year)
2.2/year1.6
(5-year)
2.6
(1-year)
Major bleeding (%)1.5
(1-year)
7.2/year1.7
(5-year)
7.9
(1-year)
ACP: Amplatzer cardiac plug, WM: Watchman, MAE: major adverse events, DRT: device-related thrombosis, PDL: peridevice leak, AF: atrial fibrillation, NA: not available.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Theofilis, P.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, K.; Tousoulis, D. Percutaneous Treatment Approaches in Atrial Fibrillation: Current Landscape and Future Perspectives. Biomedicines 2022, 10, 2268. https://doi.org/10.3390/biomedicines10092268

AMA Style

Theofilis P, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis K, Tousoulis D. Percutaneous Treatment Approaches in Atrial Fibrillation: Current Landscape and Future Perspectives. Biomedicines. 2022; 10(9):2268. https://doi.org/10.3390/biomedicines10092268

Chicago/Turabian Style

Theofilis, Panagiotis, Evangelos Oikonomou, Alexios S. Antonopoulos, Gerasimos Siasos, Konstantinos Tsioufis, and Dimitris Tousoulis. 2022. "Percutaneous Treatment Approaches in Atrial Fibrillation: Current Landscape and Future Perspectives" Biomedicines 10, no. 9: 2268. https://doi.org/10.3390/biomedicines10092268

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop