Next Article in Journal
Usefulness of Hospital Admission Chest X-ray Score for Predicting Mortality and ICU Admission in COVID-19 Patients
Next Article in Special Issue
Do Elderly Patients with Atrial Fibrillation Have Comparable Ablation Outcomes Compared to Younger Ones? Evidence from Pooled Clinical Studies
Previous Article in Journal
Upregulation of Endothelin-1 May Predict Chemotherapy-Induced Cardiotoxicity in Women with Breast Cancer
Previous Article in Special Issue
Safety and Efficacy of Left Atrial Catheter Ablation in Patients with Left Atrial Appendage Occlusion Devices
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stereotactic Radiotherapy: An Alternative Option for Refractory Ventricular Tachycardia to Drug and Ablation Therapy

1
Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
2
Department of Heart Center, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd., Chaoyang District, Beijing 100020, China
3
Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
4
Kent and Medway Medical School, Canterbury CT2 7FS, UK
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(12), 3549; https://doi.org/10.3390/jcm11123549
Submission received: 22 March 2022 / Revised: 1 June 2022 / Accepted: 15 June 2022 / Published: 20 June 2022
(This article belongs to the Special Issue Current Updates on Interventional Cardiac Electrophysiology)

Abstract

:
Refractory ventricular tachycardia (VT) often occurs in the context of organic heart disease. It is associated with significantly high mortality and morbidity rates. Antiarrhythmic drugs and catheter ablation represent the two main treatment options for refractory VT, but their use can be associated with inadequate therapeutic responses and procedure-related complications. Stereotactic body radiotherapy (SBRT) is extensively applied in the precision treatment of solid tumors, with excellent therapeutic responses. Recently, this highly precise technology has been applied for radioablation of VT, and its early results demonstrate a favorable safety profile. This review presents the potential value of SBRT in refractory VT.

1. Background

Refractory ventricular tachycardia (VT) is characterized by recurrent sustained VT despite medical or interventional treatment. Antiarrhythmic drugs have been used to treat VT for many years [1], but they are ineffective in some cases, and their use can be associated with side effects [2,3]. Catheter ablation utilizes radiofrequency or cryo-energy to destroy the scarred cardiac tissue and its surrounding tissue, effectively disrupting the re-entrant VT substrate [4,5]. Catheter ablation can be more effective than antiarrhythmic drugs [4] but may fail due to its inability to reach the arrhythmogenic target tissue or deliver adequate energy to the deep target myocardium-tissue from which the VT originates. Catheter ablation application is also limited by the occurrence of transmural injury. The recurrence rate after VT catheter ablation is as high as 50% [6]. Catheter ablation may also be associated with procedural risks and mortality [7]. An implantable cardioverter-defibrillator (ICD) can effectively terminate VT or ventricular fibrillation, reducing the risk of sudden death [8]. However, recurrent ICD shocks are painful, contribute to impaired quality of life [9,10], and are associated with higher mortality and worsening of heart failure [11,12].

2. Stereotactic Body Radiotherapy

Stereotactic body radiotherapy (SBRT) is mostly used to treat solid tumors [13]. Unlike traditional radioablation, SBRT employs radiation delivered to a 3D target volume created by various imaging technologies, accurately ablating the target volume while avoiding damage to the surrounding tissue, with up to submillimeter accuracy. Early findings reported that this noninvasive technology could effectively treat refractory VT. Unlike radiofrequency or cryo-energy ablation, SBRT is noninvasive and painless. SBRT targeting of the VT substrate of origin is an alternative for patients with refractory VT after several failed catheter ablations.

3. Mechanisms of SBRT

The exact mechanisms through which radioablation injures the target myocardium during refractory VT treatment have not been fully characterized. Some proposed radiotherapy injury mechanisms include double-strand DNA breaks that lead to cell apoptosis, vascular injury that induces tissue hypoxia, and ischemia-related cell death. Kiani [14] described the cellular effects of radiotherapy on the human myocardium. Histopathologic features were indicative of cell injury, death, and fibrosis; electron microscopy demonstrated features consistent with disruption of the cardiomyocyte architecture and cellular machinery. Indirect radiotherapy mechanisms related to tissue vasculature damage, cell hypoxia, and necrosis induce myocardial fibrosis and conduction block that may prevent further arrhythmic events [15,16].
Previous studies speculated that radioablation induced transmural fibrosis and a homogeneous myocardium, which might account for the reduced VT after SBRT. Heart radioablation with a dose of 25 Gy or higher in animal studies produced target myocardial-tissue degeneration and fibrosis and full transmural heart-injury months after the treatment [17]. Sharma et al. [18] found that radiotherapy with a single dose of at least 25 Gy could produce cavotricuspid isthmus and atrioventricular (AV) nodal blocks, and alter the electrophysiological properties at the pulmonary vein–left atrial junction. Blanck et al. [19] reported that a radioablation dose above 32.5 Gy to a healthy pig heart can induce transmural heart injury six months after treatment. Refaat et al. [20] reported that AV conduction block was induced by radioablation at a dose of 35–40 Gy, showing myocardial fibrosis in the AV node radioablation target area using immunostaining. Patients treated with a single radioablation dose of 25 Gy showed a reduced VT burden within days [21] or weeks after treatment [19,22]. Therefore, radiotherapy-induced myocardial fibrosis cannot fully explain the early effect of radioablation. An animal study found no apparent myocardial necrosis and apoptosis one month after a high-dose cardiac radioablation, further supporting the notion that fibrosis cannot explain the early antiarrhythmic effects of such a treatment [23].
Zhang et al. [24] presented evidence in an animal model and in patients that cardiac radioablation induced electrical conduction reprogramming rather than causing transmural fibrosis during the early period after treatment. Six weeks after a 25-Gy radioablation was delivered to murine hearts, no significant myocardial fibrosis or increased collagen content was detected. However, cardiac-electrophysiology remolding was found, with a significantly shortened QRS interval and increased ventricular conduction velocity. These results showed that 25-Gy radioablation induced cardiac-electrophysiology remolding without transmural fibrosis. This explains why the SBRT effect was observed within days to weeks. Western blots showed a significant increase in the expression of NaV1.5 (the cardiac voltage-gated sodium channel subunit that plays a key role in phase-0 depolarization of the action potential) and connexin 43 (a major subunit of the ventricular gap junction that helps pulse diffusion) after radioablation in murine hearts. The high expression of NaV1.5 and connexin 43 was present even 42 weeks after radioablation, achieving persistent cardiac electrical reprogramming. The Notch signaling pathway plays an important role in radioablation-induced cardiac electrophysiology reprogramming. Kim et al. [25] explored the immediate effect of radioablation on myocardial cells, showing that high-dose radioablation regulated cardiomyocyte electrophysiological activities immediately, possibly accounting for the immediate antiarrhythmic effect of SBRT on refractory VT. Additionally, animal experiments showed that radiation increased connexin 43 expression, improved the myocardial conduction velocity, reduced repolarization spatial heterogeneity, and decreased the risk of ventricular arrhythmias after myocardial infarction [26]. We can see from the above studies that radioablation-induced myocardial electrophysiology remolding, rather than fibrosis, accounted for the early effect of VT radiotherapy. The exact effects of radiation on the biology and electrophysiologic properties of the myocardium need further research.

4. SBRT Planning and Implementation

Unlike radiotherapy for tumors, the radioablation target in VT is defined by electrical properties and cannot be accurately displayed by imaging alone. Therefore, SBRT requires a multi-disciplinary team that includes oncologists, medical physicians, and cardiac electrophysiologists. Cardiac electrophysiologists can identify potential candidates and localize the re-entrant substrate using electroanatomic or other noninvasive mapping techniques. These details could assist medical physicians and oncologists to create an accurate 3D target volume, transferring these data into the SBRT system, and formulating and implementing VT radiotherapy plans.
The first VT radioablation step is to precisely identify the substrate causing the VT, often related to the underlying structural heart disease. In scar-related VT, the arrhythmia often originates from ventricular scar border zones [27]. In cases with non-ischemic causes such as dilated cardiomyopathy, the arrhythmogenic substrate often involves fibrotic areas in the myocardium and has a dispersed distribution. However, only part of the scar volume contributes to VT generation. Some surviving cardiac fibers within and around the scar tissue create slow conduction pathways and form the basis for the re-entrant VT-generating substrate. Therefore, an extensive substrate ablation that homogenizes the scar would improve the ablation success rate [28]. The myocardial scar tissue can be identified using delayed-enhanced magnetic resonance imaging (MRI), positron emission tomography–computed tomography (PET-CT), nuclear perfusion imaging, or echocardiography. Most patients with refractory VT have undergone 3D electroanatomic mapping and an unsuccessful catheter ablation procedure before cardiac SBRT. Noninvasive arrhythmia substrate mapping before SBRT would help define and localize the ablation targets [29].
Precise targeting of the arrhythmogenic substrate is crucial in VT radioablation, as it could help reduce off-target ablation of surrounding healthy myocardium, organs, and implantable electronic devices. Accurately transforming the substrate information to target volume planning is a difficult task. Multimodal integration of the electroanatomic mapping with computed tomography (CT) planning could help accurately localize the target on the CT images [30]. High-quality endocardial-surface and -chamber mapping could help to precisely merge the information with cardiac CT [31]. The RAVENTA trial found remarkable differences between the electroanatomic mapping outcomes and target volume planning during VT radioablation [32]. Therefore, highly effective and uniform standards or guidelines are required to transfer the electroanatomic mapping information to the CT planning.
The heart is a moving target due to its intrinsic spontaneous mechanical activity and motions caused by respiration. Initially, SBRT was thought to be unsuitable for cardiac arrhythmia treatment due to difficulties in localizing a moving target. However, with the great progress in imaging technologies such as the gating and tracking systems, the scope of SBRT has been expanded to include VT treatment [22]. Imaging strategies are used to differentiate the target tissue volume from the surrounding healthy tissue. Motion compensation strategies include dampening or inhibition to restrain respiratory motion, gating to release radiation at a specific time of the respiratory cycle, and tracking to make the radiation beam follow the moving target [33]. Ho et al. [34] reported that computational ECG mapping and a protocol-guided respiratory gating system could improve target volume planning precision, helping with SBRT planning and implementation. Cha et al. [35] proposed a new method, using deep-inspiration breath-hold to separate the target from the stomach, reducing the dose delivered to the gastrointestinal tract during cardiac SBRT. Real-time cardiorespiratory-motion-mitigated MRI could help compensate for cardiac and respiratory motions simultaneously [36]. Although many techniques have been used to minimize injury to the surrounding tissues, it is impossible to fully avoid the off-target delivery of radiation beams due to the respiratory and cardiac motions.
SBRT duration ranges between 10 and 90 min, depending on the radiation dose, the machines used, and the target tissue volume, location, and size (Table 1). Another key issue in SBRT for VT ablation is the radiation dose. Blanck et al. [19] showed, in a pig model, that a dose of 17.5 Gy did not induce any significant fibrosis in pulmonary veins, a dose of 25 Gy induced mild fibrosis, and a dose above 32.5 Gy might lead to transmural fibrosis. Animal study outcomes supported 25 Gy as the radioablation threshold dosage to create target myocardial-tissue fibrosis, but higher radiation dosages (35–40 Gy) increased the risks of radiation-related complications [18,19,37]. Based on preclinical results and experience in oncology SBRT, most clinical studies used 25 Gy as the basic treatment dose. The currently prescribed 25-Gy dose might be insufficient to form a homogenous transmural scar. Zhang et al. [24] proposed that the radioablation mechanism was mediated by electrophysiologic reprogramming rather than myocardial fibrosis. Using a dose de-escalation method, they found that a dose of 5–10 Gy did not induce significant electrophysiologic reprogramming, while a dose higher than 15 Gy increased the ventricular conduction velocity and shortened the QRS interval. The largest electrophysiologic effects were detected at a dose of 25 Gy. Elucidation of the optimal dose regimen needs further study.

5. SBRT for VT Ablation: Clinical Experience

To date, a limited number of prospective and retrospective studies or case series focusing on the clinical evidence of SBRT for VT have been published. Loo et al. [38] delivered the first in-human cardiac arrhythmia treatment in 2012 using stereotactic radioablation with a radiation dose of 25 Gy. After a 2-month blanking period, the number of VT episodes remained low for seven months, with no acute or late complications. However, refractory VT occurred nine months after the procedure in the context of an exacerbation of chronic obstructive pulmonary disease, culminating in the patient’s death.
In a case series reported by Cuculich et al. [22], five patients at high risk of refractory VT underwent SBRT with a single delivery dose of 25 Gy and a 14-min mean radioablation duration. The pre-procedural VT burden of these patients was 6577 episodes over a 3-month time period. During the 6-week post-ablation blanking period, the VT episodes decreased to 680 and declined to 4 over the next 46 patient-months. All five patients presented with a significant reduction in VT burden from the baseline. The mean left-ventricular-ejection fraction was preserved throughout the follow-up.
In 2019, Robinson et al. [40] published a phase I-II prospective trial of VT radioablation. Nineteen patients with treatment-refractory VT or premature-ventricular-contraction (PVC) cardiomyopathy were enrolled and received noninvasive electrophysiology-guided cardiac radioablation with a single delivery of 25 Gy. The number of VT episodes decreased from 119 to 3 in 18 patients after the ablation, 17 of which (94%) presented with reduced VT or PVC burden. The overall 6- and 12-month survival rates were 89% and 72%, respectively; the dual antiarrhythmic drug use declined from 59 to 12%; and the quality of life at six months improved in five of the nine Short Form-36 domains. Evidently, noninvasive electrophysiology-guided cardiac radioablation could bring favorable clinical benefits to patients with VT or PVC-related cardiomyopathy.
Neuwirth et al. [39] reported a case series of ten patients with structural heart disease and refractory VT. They delivered 25 Gy to the planning target volumes using CyberKnife. Compared to the baseline, the VT burden significantly decreased by 87.5% during the 28-month follow-up. Three patients died of non-arrhythmic causes. After a blanking period of 90 days, eight of the patients experienced a recurrence of VT. The mean times from treatment to the first antitachycardia pacing and ICD shock were 6.5 and 21 months, respectively. The follow-up results showed that SBRT failed in two patients, and two patients showed a delayed response of three and six months, respectively. One patient with previously known mitral regurgitation showed progression of the regurgitation and valvular morphology changes 17 months after the radioablation procedure. This study showed that SBRT was a safe and effective long-term treatment for refractory VT and could improve the survival rate of these patients.
Gianni et al. [42] described five patients with structural heart disease and refractory VT. A single-fraction radiation dose of 25 Gy was delivered to the target volume using CyberKnife at a mean treatment duration of 82 min. Four of the five patients presented with significant reductions in the number of VT episodes and in the use of antiarrhythmic drugs during the first six months after the ablation. All five patients experienced VT recurrence during the 12-month follow-up, of which two died from heart failure worsening. Those researchers found no evidence that the radioablation injured the cardiac tissue around the target volume. Despite the initial promising results, SBRT did not show long-term effectiveness in patients with refractory VT. No radiation-related complications were noted during follow-up, suggesting an acceptable safety profile for SBRT. Further studies are needed to elucidate the mechanisms associated with the lack of long-term efficacy.
Lloyd et al. [41] reported ten patients with advanced heart failure (four with ischemic and six with non-ischemic cardiomyopathy) and refractory VT who underwent SBRT. The mean patient age was 61 years. The results showed that the number of VT episodes decreased by 69%, antitachycardia pacing sequences decreased by 48%, and ICD shocks decreased by 68% after the SBRT. One patient in this study did not respond to the SBRT. After excluding this patient, a significant decrease in the VT burden was observed after treatment.
Chin et al. [43] retrospectively analyzed eight consecutive patients with refractory VT who underwent SBRT, with an average age of 75 years and a mean ejection fraction of 21%. All patients were male, half with ischemic and half with non-ischemic cardiomyopathy. A mean dose of 22.2 Gy was delivered to the planning target volume during a single radioablation treatment session. During the 7.8-month follow-up, the number of ICD shocks decreased from 69.5 to 13.3. No acute complications or definite peripheral organ toxicities occurred during follow-up. Three patients died during follow-up, of causes unrelated to the SBRT. This patient group experienced VT burden reduction after the SBRT, but, unlike previous reports, they showed no immediate effects.
Carbucicchio et al. [44] presented the preliminary results of their spontaneous, prospective, single-arm, phase Ib/II single-center STRA-MI-VT Study. The study investigated the safety and efficacy of SBRT in eight patients with intractable VT. Of the seven patients who underwent SBRT (mean age, 70 ± 7 years; ejection fraction, 27 ± 11%), three had ischemic and four had non-ischemic cardiomyopathy. No treatment-related serious adverse events occurred at a median follow-up of eight months. Three patients died from non-SBRT-related causes, while four completed the 6-month follow-up. The number of VT episodes decreased from 29 to 11 and 2, three and six months post-treatment, respectively. The respective number of ICD shocks decreased from 11 to 0 and 2. All patients showed a significant reduction in the number of VT episodes and no electrical storm recurrence six months after treatment. The STRA-MI-VT Study showed excellent short-term VT radioablation treatment outcomes but showed no long-term effects.
The study by Qian et al. [45] aimed to determine the feasibility of using radioablation on the arrhythmogenic substrate of myocardial scars in patients with ischemic cardiomyopathy. Six male patients with ischemic cardiomyopathy and refractory VT underwent SBRT targeting of the extensive scar substrate. The median planning target volume was 319 mL, and the radioablation dose was 25 Gy. ICD-treated or sustained VT episodes did not decrease significantly after SBRT, but ICD shocks decreased significantly from 12 to 0. Three patients died of heart failure during the 7.7-month follow-up period, and three experienced complications, including heart failure exacerbation, pneumonia, and asymptomatic pericardial effusion. Repeat catheter ablation was performed in four patients at 32, 167, 288, and 396 days post-SBRT. This study suggested that SBRT-induced substrate modifications did not significantly reduce the VT burden in patients with ischemic cardiomyopathy, similar to a recent report demonstrating that radioablation did not have long-term effectiveness in patients with serious ischemic cardiomyopathy [38]. Further study is warranted to evaluate the radiobiology of myocardial scars, optimal radiation dose, target location, and the effectiveness and safety of refractory VT radioablation in patients with ischemic cardiomyopathy.
Ho et al. [34] enrolled six consecutive patients with refractory VT managed by SBRT. The VT origin sites were identified noninvasively by a 3D computational electrocardiogram (ECG) algorithm and compared to available electroanatomic maps. A 25-Gy radioablation was delivered to the target at the end of expiration. Respiratory gating facilitated small planning target volumes and prevented gastrointestinal complications. ICD shocks decreased from 23 to 0.67 per patient six months post-SRBT. This study showed that a workflow that combined computational ECG mapping and protocol-guided respiratory gating was effective and safe, and could improve SRBT planning.
However, long-term follow-ups indicate a high VT recurrence rate in patients with refractory VT after SRBT, the reasons for which remain unknown. Gianni et al. [42] described three patients with recurrent VT after radioablation. Voltage mapping during repeated radiofrequency catheter ablation showed remaining low voltage, fractionated electrograms within the radioablation target regions. Some surviving cardiomyocytes in the scar suggested incomplete homogenization. Therefore, the 25-Gy radiation dose used in previous studies might be insufficient to destroy the entire target tissue. The uniform delivery of a 25-Gy radiation dose, regardless of the planning target volume, size, and characteristics, needs further exploration and study. Another possibility is that the identified substrate might not be very accurate, as location discrepancies were introduced when the data were transferred to the CT imaging system [32]. Furthermore, aggravation of the myocardial ischemia or exacerbation of the heart failure might produce a new VT substrate, especially in patients with severe ischemic cardiomyopathy. The findings of the many ongoing clinical trials will be able to address some of these issues.

6. Safety and Complications

Several studies have reported that the VT radioablation treatment was safe during short-to-medium follow-ups, with only a few complications reported. These included pericarditis (n = 1), the progression of valvular disease (n = 1), self-resolving pneumonitis (n = 2), and delayed pericardial effusion (n = 5) [39,40]. There have been no reports of VT radioablation treatment as the direct cause of death (Table 1). Longer-term results from the Phase I/II ENCORE-VT Study showed one serious Grade 4 complication (gastro-pericardial fistula) at 2.4 years requiring surgical repair [52]. Haskova et al. [53] reported a refractory VT patient who died because of a bleeding esophago-pericardial fistula six months after a 25-Gy radioablation. To date, not many patients have undergone SBRT, so two fistula cases suggest that this fatal complication is not uncommon. Nevertheless, we may gain some insights from cancer radiotherapy. Several studies showed a significant relationship between the delivered cardiac radiation doses and the long-term (several years or even decades) cumulative incidence of cardiac events in the coronary arteries, conduction system, valvular structures, myocardium, and pericardium [54]. We need to be alert to late complications such as esophago-pericardial fistula. Long-term follow-up is needed to fully identify potential late complications.

7. Limitations

The following limitations of SBRT should be taken into consideration. First, it requires accurate identification and localization of the substrate. Second, an optimal radiation dosing regimen is still lacking and should be further elucidated. Third, the long-term safety and efficacy of SBRT remain unknown. Therefore, SBRT should be reserved for patients with refractory VT following failed medical therapy and ablation.

8. Conclusions

SBRT represents a noninvasive option for cases of refractory VT with failed antiarrhythmic therapy and ablation. Further clinical trials and registry studies are needed to better inform and optimize its treatment parameters and characterize its long-term safety.

Author Contributions

Conceptualization, G.X., T.L.; writing—original draft preparation, W.S., G.X. and X.W.; writing—review and editing, W.S., N.Z., X.L. and G.T.; supervision, G.L. and T.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from the National Natural Science Foundation of China (No. 82000313 to W.S., No. 81970270 to T.L.), Tianjin Natural Science Foundation (No. 18JCYBJC92700 to G.X), and Tianjin Key Medical Discipline (Specialty) Construction Project.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Bazoukis, G.; Tse, G.; Letsas, K.P.; Thomopoulos, C.; Naka, K.K.; Korantzopoulos, P.; Bazoukis, X.; Michelongona, P.; Papadatos, S.S.; Vlachos, K.; et al. Impact of ranolazine on ventricular arrhythmias—A systematic review. J. Arrhythmia 2018, 34, 124–128. [Google Scholar] [CrossRef] [PubMed]
  2. Antiarrhythmics versus Implantable Defibrillators Investigators. A comparison of antiarrhythmic-drug therapy with implantable defibrillators in patients resuscitated from near-fatal ventricular arrhythmias. N. Engl. J. Med. 1997, 337, 1576–1583. [Google Scholar] [CrossRef] [PubMed]
  3. Connolly, S.J.; Dorian, P.; Roberts, R.S.; Gent, M.; Bailin, S.; Fain, E.S.; Thorpe, K.; Champagne, J.; Talajic, M.; Coutu, B.; et al. Comparison of beta-blockers, amiodarone plus beta-blockers, or sotalol for prevention of shocks from implantable cardioverter defibrillators: The optic study: A randomized trial. JAMA 2006, 295, 165–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sapp, J.L.; Wells, G.A.; Parkash, R.; Stevenson, W.G.; Blier, L.; Sarrazin, J.F.; Thibault, B.; Rivard, L.; Gula, L.; Leong-Sit, P.; et al. Ventricular tachycardia ablation versus escalation of antiarrhythmic drugs. N. Engl. J. Med. 2016, 375, 111–121. [Google Scholar] [CrossRef]
  5. Rivera, S.; Ricapito, M.D.; Tomas, L.; Parodi, J.; Bardera Molina, G.; Banega, R.; Bueti, P.; Orosco, A.; Reinoso, M.; Caro, M.; et al. Results of cryoenergy and radiofrequency-based catheter ablation for treating ventricular arrhythmias arising from the papillary muscles of the left ventricle, guided by intracardiac echocardiography and image integration. Circ. Arrhythmia Electrophysiol. 2016, 9, e003874. [Google Scholar] [CrossRef] [Green Version]
  6. Piers, S.R.; Leong, D.P.; van Taxis, C.F.; Tayyebi, M.; Trines, S.A.; Pijnappels, D.A.; Delgado, V.; Schalij, M.J.; Zeppenfeld, K. Outcome of ventricular tachycardia ablation in patients with nonischemic cardiomyopathy: The impact of noninducibility. Circ. Arrhythmia Electrophysiol. 2013, 6, 513–521. [Google Scholar] [CrossRef] [Green Version]
  7. Santangeli, P.; Muser, D.; Maeda, S.; Filtz, A.; Zado, E.S.; Frankel, D.S.; Dixit, S.; Epstein, A.E.; Callans, D.J.; Marchlinski, F.E. Comparative effectiveness of antiarrhythmic drugs and catheter ablation for the prevention of recurrent ventricular tachycardia in patients with implantable cardioverter-defibrillators: A systematic review and meta-analysis of randomized controlled trials. Heart Rhythm 2016, 13, 1552–1559. [Google Scholar] [CrossRef]
  8. Al-Khatib, S.M.; Stevenson, W.G.; Ackerman, M.J.; Bryant, W.J.; Callans, D.J.; Curtis, A.B.; Deal, B.J.; Dickfeld, T.; Field, M.E.; Fonarow, G.C.; et al. 2017 aha/acc/hrs guideline for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: Executive summary: A report of the american college of cardiology/american heart association task force on clinical practice guidelines and the heart rhythm society. Heart Rhythm 2018, 15, e190–e252. [Google Scholar]
  9. Schron, E.B.; Exner, D.V.; Yao, Q.; Jenkins, L.S.; Steinberg, J.S.; Cook, J.R.; Kutalek, S.P.; Friedman, P.L.; Bubien, R.S.; Page, R.L.; et al. Quality of life in the antiarrhythmics versus implantable defibrillators trial: Impact of therapy and influence of adverse symptoms and defibrillator shocks. Circulation 2002, 105, 589–594. [Google Scholar] [CrossRef] [Green Version]
  10. Perini, A.P.; Kutyifa, V.; Veazie, P.; Daubert, J.P.; Schuger, C.; Zareba, W.; McNitt, S.; Rosero, S.; Tompkins, C.; Padeletti, L.; et al. Effects of implantable cardioverter/defibrillator shock and antitachycardia pacing on anxiety and quality of life: A madit-rit substudy. Am. Heart J. 2017, 189, 75–84. [Google Scholar] [CrossRef]
  11. Bazoukis, G.; Tse, G.; Korantzopoulos, P.; Liu, T.; Letsas, K.P.; Stavrakis, S.; Naka, K.K. Impact of implantable cardioverter-defibrillator interventions on all-cause mortality in heart failure patients: A meta-analysis. Cardiol. Rev. 2019, 27, 160–166. [Google Scholar] [CrossRef] [PubMed]
  12. Poole, J.E.; Johnson, G.W.; Hellkamp, A.S.; Anderson, J.; Callans, D.J.; Raitt, M.H.; Reddy, R.K.; Marchlinski, F.E.; Yee, R.; Guarnieri, T.; et al. Prognostic importance of defibrillator shocks in patients with heart failure. N. Engl. J. Med. 2008, 359, 1009–1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Potters, L.; Kavanagh, B.; Galvin, J.M.; Hevezi, J.M.; Janjan, N.A.; Larson, D.A.; Mehta, M.P.; Ryu, S.; Steinberg, M.; Timmerman, R.; et al. American society for therapeutic radiology and oncology (astro) and american college of radiology (acr) practice guideline for the performance of stereotactic body radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 2010, 76, 326–332. [Google Scholar] [CrossRef] [PubMed]
  14. Kiani, S.; Kutob, L.; Schneider, F.; Higgins, K.A.; Lloyd, M.S. Histopathologic and ultrastructural findings in human myocardium after stereotactic body radiation therapy for recalcitrant ventricular tachycardia. Circ. Arrhythmia Electrophysiol. 2020, 13, e008753. [Google Scholar] [CrossRef] [PubMed]
  15. Song, C.W.; Lee, Y.J.; Griffin, R.J.; Park, I.; Koonce, N.A.; Hui, S.; Kim, M.S.; Dusenbery, K.E.; Sperduto, P.W.; Cho, L.C. Indirect tumor cell death after high-dose hypofractionated irradiation: Implications for stereotactic body radiation therapy and stereotactic radiation surgery. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 166–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Song, C.W.; Glatstein, E.; Marks, L.B.; Emami, B.; Grimm, J.; Sperduto, P.W.; Kim, M.S.; Hui, S.; Dusenbery, K.E.; Cho, L.C. Biological principles of stereotactic body radiation therapy (sbrt) and stereotactic radiation surgery (srs): Indirect cell death. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 21–34. [Google Scholar] [CrossRef]
  17. Lehmann, H.I.; Deisher, A.J.; Takami, M.; Kruse, J.J.; Song, L.; Anderson, S.E.; Cusma, J.T.; Parker, K.D.; Johnson, S.B.; Asirvatham, S.J.; et al. External Arrhythmia Ablation Using Photon Beams: Ablation of the Atrioventricular Junction in an Intact Animal Model. Circ. Arrhythmia Electrophysiol. 2017, 10, e004304. [Google Scholar] [CrossRef]
  18. Sharma, A.; Wong, D.; Weidlich, G.; Fogarty, T.; Jack, A.; Sumanaweera, T.; Maguire, P. Noninvasive stereotactic radiosurgery (cyberheart) for creation of ablation lesions in the atrium. Heart Rhythm 2010, 7, 802–810. [Google Scholar] [CrossRef]
  19. Blanck, O.; Bode, F.; Gebhard, M.; Hunold, P.; Brandt, S.; Bruder, R.; Grossherr, M.; Vonthein, R.; Rades, D.; Dunst, J. Dose-escalation study for cardiac radiosurgery in a porcine model. Int. J. Radiat. Oncol. Biol. Phys. 2014, 89, 590–598. [Google Scholar] [CrossRef]
  20. Refaat, M.M.; Ballout, J.A.; Zakka, P.; Hotait, M.; Al Feghali, K.A.; Gheida, I.A.; Saade, C.; Hourani, M.; Geara, F.; Tabbal, M.; et al. Swine atrioventricular node ablation using stereotactic radiosurgery: Methods and in vivo feasibility investigation for catheter-free ablation of cardiac arrhythmias. J. Am. Heart Assoc. 2017, 6, e007193. [Google Scholar] [CrossRef] [Green Version]
  21. Jumeau, R.; Ozsahin, M.; Schwitter, J.; Vallet, V.; Duclos, F.; Zeverino, M.; Moeckli, R.; Pruvot, E.; Bourhis, J. Rescue procedure for an electrical storm using robotic non-invasive cardiac radio-ablation. Radiother. Oncol. 2018, 128, 189–191. [Google Scholar] [CrossRef] [PubMed]
  22. Cuculich, P.S.; Schill, M.R.; Kashani, R.; Mutic, S.; Lang, A.; Cooper, D.; Faddis, M.; Gleva, M.; Noheria, A.; Smith, T.W.; et al. Noninvasive cardiac radiation for ablation of ventricular tachycardia. N. Engl. J. Med. 2017, 377, 2325–2336. [Google Scholar] [CrossRef] [PubMed]
  23. Cha, M.J.; Seo, J.W.; Kim, H.J.; Kim, M.K.; Yoon, H.S.; Jo, S.W.; Oh, S.; Chang, J.H. Early Changes in Rat Heart After High-Dose Irradiation: Implications for Antiarrhythmic Effects of Cardiac Radioablation. J. Am. Heart Assoc. 2021, 10, e019072. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, D.M.; Navara, R.; Yin, T.; Szymanski, J.; Goldsztejn, U.; Kenkel, C.; Lang, A.; Mpoy, C.; Lipovsky, C.E.; Qiao, Y.; et al. Cardiac radiotherapy induces electrical conduction reprogramming in the absence of transmural fibrosis. Nat. Commun. 2021, 12, 5558. [Google Scholar] [CrossRef]
  25. Kim, J.S.; Choi, S.W.; Park, Y.G.; Kim, S.J.; Choi, C.H.; Cha, M.J.; Chang, J.H. Impact of High-Dose Irradiation on Human iPSC-Derived Cardiomyocytes Using Multi-Electrode Arrays: Implications for the Antiarrhythmic Effects of Cardiac Radioablation. Int. J. Mol. Sci. 2021, 23, 351. [Google Scholar] [CrossRef]
  26. Amino, M.; Yoshioka, K.; Tanabe, T.; Tanaka, E.; Mori, H.; Furusawa, Y.; Zareba, W.; Yamazaki, M.; Nakagawa, H.; Honjo, H.; et al. Heavy ion radiation up-regulates Cx43 and ameliorates arrhythmogenic substrates in hearts after myocardial infarction. Cardiovasc. Res. 2006, 7, 412–421. [Google Scholar] [CrossRef]
  27. Aliot, E.M.; Stevenson, W.G.; Almendral-Garrote, J.M.; Bogun, F.; Calkins, C.H.; Delacretaz, E.; Bella, P.D.; Hindricks, G.; Jaïs, P.; Josephson, M.E.; et al. EHRA/HRS expert consensus on catheter ablation of ventricular arrhythmias: Developed in a partnership with the European heart rhythm association (EHRA), a registered branch of the European society of cardiology (ESC), and the heart rhythm society (HRS); in collaboration with the American college of cardiology (ACC) and the American heart association (AHA). Heart Rhythm 2009, 6, 886–933. [Google Scholar]
  28. Di Biase, L.; Burkhardt, J.D.; Lakkireddy, D.; Carbucicchio, C.; Mohanty, S.; Mohanty, P.; Trivedi, C.; Santangeli, P.; Bai, R.; Forleo, G.; et al. Ablation of stable VTs versus substrate ablation in ischemic cardiomyopathy: The vista randomized multicenter trial. J. Am. Coll. Cardiol. 2015, 66, 2872–2882. [Google Scholar] [CrossRef] [Green Version]
  29. Kim, E.J.; Davogustto, G.; Stevenson, W.G.; John, R.M. Non-invasive cardiac radiation for ablation of ventricular tachycardia: A new therapeutic paradigm in electrophysiology. Arrhythmia Electrophysiol. Rev. 2018, 7, 8–10. [Google Scholar] [CrossRef]
  30. Hohmann, S.; Henkenberens, C.; Zormpas, C.; Christiansen, H.; Bauersachs, J.; Duncker, D.; Veltmann, C. A novel open-source software-based high-precision workflow for target definition in cardiac radioablation. J. Cardiovasc. Electrophysiol. 2020, 31, 2689–2695. [Google Scholar] [CrossRef]
  31. Abdel-Kafi, S.; Sramko, M.; Omara, S.; de Riva, M.; Cvek, J.; Peichl, P.; Kautzner, J.; Zeppenfeld, K. Accuracy of electroanatomical mapping-guided cardiac radiotherapy for ventricular tachycardia: Pitfalls and solutions. Europace 2021, 23, 1989–1997. [Google Scholar] [CrossRef] [PubMed]
  32. Boda-Heggemann, J.; Blanck, O.; Mehrhof, F.; Ernst, F.; Buergy, D.; Fleckenstein, J.; Tülümen, E.; Krug, D.; Siebert, F.A.; Zaman, A.; et al. Interdisciplinary clinical target volume generation for cardiac radioablation: Multicenter benchmarking for the radiosurgery for ventricular tachycardia (raventa) trial. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 745–756. [Google Scholar] [CrossRef]
  33. Timmerman, R.D.; Kavanagh, B.D.; Cho, L.C.; Papiez, L.; Xing, L. Stereotactic body radiation therapy in multiple organ sites. J. Clin. Oncol. 2007, 25, 947–952. [Google Scholar] [CrossRef] [PubMed]
  34. Ho, G.; Atwood, T.F.; Bruggeman, A.R.; Moore, K.L.; McVeigh, E.; Villongco, C.T.; Han, F.T.; Hsu, J.C.; Hoffmayer, K.S.; Raissi, F.; et al. Computational ECG mapping and respiratory gating to optimize stereotactic ablative radiotherapy workflow for refractory ventricular tachycardia. Heart Rhythm O2 2021, 2, 511–520. [Google Scholar] [CrossRef] [PubMed]
  35. Cha, M.J.; Cuculich, P.S.; Robinson, C.G.; Chang, J.H. Tailored stereotactic radiotherapy technique using deep inspiration breath-hold to reduce stomach dose for cardiac radioablation. Radiat. Oncol. J. 2021, 39, 167–173. [Google Scholar] [CrossRef]
  36. Akdag, O.; Borman, P.T.; Woodhead, P.; Uijtewaal, P.; Mandija, S.; Van Asselen, B.; Verhoeff, J.J.; Raaymakers, B.W.; Fast, M.F. First experimental exploration of real-time cardiorespiratory motion management for future stereotactic arrhythmia radioablation treatments on the MR-linac. Phys. Med. Biol. 2022, 67, 065003. [Google Scholar] [CrossRef]
  37. Zei, P.C.; Wong, D.; Gardner, E.; Fogarty, T.; Maguire, P. Safety and efficacy of stereotactic radioablation targeting pulmonary vein tissues in an experimental model. Heart Rhythm 2018, 15, 1420–1427. [Google Scholar] [CrossRef]
  38. Loo, B.W., Jr.; Soltys, S.G.; Wang, L.; Lo, A.; Fahimian, B.P.; Iagaru, A.; Norton, L.; Shan, X.; Gardner, E.; Fogarty, T.; et al. Stereotactic ablative radiotherapy for the treatment of refractory cardiac ventricular arrhythmia. Circ. Arrhythmia Electrophysiol. 2015, 8, 748–750. [Google Scholar] [CrossRef] [Green Version]
  39. Neuwirth, R.; Cvek, J.; Knybel, L.; Jiravsky, O.; Molenda, L.; Kodaj, M.; Fiala, M.; Peichl, P.; Feltl, D.; Januška, J.; et al. Stereotactic radiosurgery for ablation of ventricular tachycardia. Europace 2019, 21, 1088–1095. [Google Scholar] [CrossRef]
  40. Robinson, C.G.; Samson, P.P.; Moore, K.M.; Hugo, G.D.; Knutson, N.; Mutic, S.; Goddu, S.M.; Lang, A.; Cooper, D.H.; Faddis, M.; et al. Phase i/ii trial of electrophysiology-guided noninvasive cardiac radioablation for ventricular tachycardia. Circulation 2019, 139, 313–321. [Google Scholar] [CrossRef]
  41. Lloyd, M.S.; Wight, J.; Schneider, F.; Hoskins, M.; Attia, T.; Escott, C.; Lerakis, S.; Higgins, K.A. Clinical experience of stereotactic body radiation for refractory ventricular tachycardia in advanced heart failure patients. Heart Rhythm 2020, 17, 415–422. [Google Scholar] [CrossRef]
  42. Gianni, C.; Rivera, D.; Burkhardt, J.D.; Pollard, B.; Gardner, E.; Maguire, P.; Zei, P.C.; Natale, A.; Al-Ahmad, A. Stereotactic arrhythmia radioablation for refractory scar-related ventricular tachycardia. Heart Rhythm 2020, 17, 1241–1248. [Google Scholar] [CrossRef]
  43. Chin, R.; Hayase, J.; Hu, P.; Cao, M.; Deng, J.; Ajijola, O.; Do, D.; Vaseghi, M.; Buch, E.; Khakpour, H.; et al. Non-invasive stereotactic body radiation therapy for refractory ventricular arrhythmias: An institutional experience. J. Interv. Cardiol. Electrophysiol. 2021, 61, 535–543. [Google Scholar] [CrossRef]
  44. Carbucicchio, C.; Andreini, D.; Piperno, G.; Catto, V.; Conte, E.; Cattani, F.; Bonomi, A.; Rondi, E.; Piccolo, C.; Vigorito, S.; et al. Stereotactic radioablation for the treatment of ventricular tachycardia: Preliminary data and insights from the STRA-MI-VT phase Ib/II study. J. Interv. Cardiol. Electrophysiol. 2021, 62, 427–439. [Google Scholar] [CrossRef]
  45. Qian, P.C.; Quadros, K.; Aguilar, M.; Wei, C.; Boeck, M.; Bredfeldt, J.; Cochet, H.; Blankstein, R.; Mak, R.; Sauer, W.H.; et al. Substrate Modification Using Stereotactic Radioablation to Treat Refractory Ventricular Tachycardia in Patients With Ischemic Cardiomyopathy. JACC Clin. Electrophysiol. 2022, 8, 49–58. [Google Scholar] [CrossRef]
  46. Haskova, J.; Peichl, P.; Pirk, J.; Cvek, J.; Neuwirth, R.; Kautzner, J. Stereotactic radiosurgery as a treatment for recurrent ventricular tachycardia associated with cardiac fibroma. HeartRhythm Case Rep. 2019, 5, 44–47. [Google Scholar] [CrossRef] [Green Version]
  47. Marti-Almor, J.; Jimenez-Lopez, J.; Rodriguez de Dios, N.; Tizon, H.; Valles, E.; Algara, M. Noninvasive ablation of ventricular tachycardia with stereotactic radiotherapy in a patient with arrhythmogenic right ventricular cardiomyopathy. Rev. Esp. Cardiol. (Engl. Ed). 2020, 73, 97–99. [Google Scholar] [CrossRef]
  48. Scholz, E.P.; Seidensaal, K.; Naumann, P.; Andre, F.; Katus, H.A.; Debus, J. Risen from the dead: Cardiac stereotactic ablative radiotherapy as last rescue in a patient with refractory ventricular fibrillation storm. HeartRhythm Case Rep. 2019, 5, 329–332. [Google Scholar] [CrossRef] [Green Version]
  49. Zeng, L.J.; Huang, L.H.; Tan, H.; Zhang, H.C.; Mei, J.; Shi, H.F.; Jiang, C.Y.; Tan, C.; Zheng, J.W.; Liu, X.P. Stereotactic body radiation therapy for refractory ventricular tachycardia secondary to cardiac lipoma: A case report. Pacing Clin. Electrophysiol. 2019, 42, 1276–1279. [Google Scholar] [CrossRef]
  50. Krug, D.; Blanck, O.; Demming, T.; Dottermusch, M.; Koch, K.; Hirt, M.; Kotzott, L.; Zaman, A.; Eidinger, L.; Siebert, F.A.; et al. Stereotactic body radiotherapy for ventricular tachycardia (cardiac radiosurgery): First-in-patient treatment in Germany. Strahlenther. Onkol. 2020, 196, 23–30. [Google Scholar] [CrossRef]
  51. Mayinger, M.; Kovacs, B.; Tanadini-Lang, S.; Ehrbar, S.; Wilke, L.; Chamberlain, M.; Moreira, A.; Weitkamp, N.; Brunckhorst, C.; Duru, F.; et al. First magnetic resonance imaging-guided cardiac radioablation of sustained ventricular tachycardia. Radiother. Oncol. 2020, 152, 203–207. [Google Scholar] [CrossRef] [PubMed]
  52. Robinson, C.G.; Samson, P.; Moore, K.M.S.; Hugo, G.D.; Knutson, N.; Mutic, S. Longer Term Results from a Phase I/II Study of EP-guided Noninvasive Cardiac Radioablation for Treatment of Ventricular Tachycardia (ENCORE-VT). Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 682. [Google Scholar] [CrossRef]
  53. Haskova, J.; Jedlickova, K.; Cvek, J.; Knybel, L.; Neuwirth, R.; Kautzner, J. Oesophagopericardial fistula as a late complication of stereotactic radiotherapy for recurrent ventricular tachycardia. Europace 2022, euab326. [Google Scholar] [CrossRef]
  54. Bergom, C.; Bradley, J.A.; Ng, A.K.; Samson, P.; Robinson, C.; Lopez-Mattei, J.; Mitchell, J.D. Past, Present, and Future of Radiation-Induced Cardiotoxicity: Refinements in Targeting, Surveillance, and Risk Stratification. JACC CardioOncol. 2021, 3, 343–359. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of the different studies on stereotactic radiotherapy for ventricular arrhythmia.
Table 1. Summary of the different studies on stereotactic radiotherapy for ventricular arrhythmia.
Study YearPatient NumberSexMean Age (Years)Type of CMPLVEF (Mean, %)Dose (Gy)PTV (Mean, mL)Treatment Time (Mean, Min)Delay for EfficacyFollow-Up (Months)Complications
Loo et al. [38] 20151M71ICMP2425-90After 2 months9Died from COPD exacerbation at month 9
Cuculich et al. [22] 201754 M;
1 F
662 ICMP;
3 NICMP
23254914Progressive effect after ablation, but maximum effect after 6 weeks12One fatal stroke 3 weeks after treatment
Jumeau et al. [21] 20181M75NICMP30252145Immediate4None
Neuwirth et al. [39] 2019109 M;
1 F
668 ICMP;
2 NICMP
272522.268Progressive effect28Three died of non-arrhythmic causes; progression of mitral valve regurgitation at 17 months
Robinson et al. [40] 20191917 M;
2 F
6611 ICMP;
5 NICMP;
3 others
252598.915Within the first 6 weeks13Pericarditis; heart failure exacerbation at 2 months
Lloyd et al. [41] 2020107 M;
3 F
624 ICMP;
4 NICMP;
2 others
-2581.4-Within the first 2 weeks6Mild pneumonitis responsive to corticosteroids in two patients
Gianni et al. [42] 202055 M634 ICMP;
1 NICMP
342514382Four patients had marked reduction in VT burden during first 6 months12Two died of heart failure
Chin et al. [43] 202188 M754 ICMP;
4 NICMP
2122.2121.418.23 months7.8No acute complications, three patient deaths in the follow-up period, unrelated to SBRT.
Carbucicchio C [44] 202178 M703 ICMP;
4 NICMP
2725183313 months8three patient deaths in the follow-up period, unrelated to SBRT.
Qian et al. [45] 202166 M726 ICMP2025319-6 months7.73 patients died of heart failure; 3 of 6 patients had possible adverse events
Ho et al. [34] 202166 M742 ICMP;
4 NICMP
2925120.321.1-61 patient Pericardial effusion 12 months after therapy
Haskova et al. [46] 20181-34NICMP-25--8 months8-
Martí Almor et al. [47] 20201M64NICMP-25-4Immediate4None
Scholz et al. [48] 20191M53ICMP-3082.45/302 weeks2None
Zeng et al. [49] 20191M29NICMP-2471-1 month4None
Krug et al. [50] 20191M78NICMP152542.215Days-Died 57 days after ablation due to sepsis-associated cardiac circulatory failure
Mayinger et al. [51] 20201M71NICMP2525115.12448 h3None
CMP—cardiomyopathy; COPD—chronic obstructive pulmonary disease; F—female; PTV—planning target volume; ICMP—ischemic cardiomyopathy; LVEF—left ventricular ejection fraction; M—male; NICMP—non-ischemic cardiomyopathy; VT—ventricular tachycardia.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shangguan, W.; Xu, G.; Wang, X.; Zhang, N.; Liu, X.; Li, G.; Tse, G.; Liu, T. Stereotactic Radiotherapy: An Alternative Option for Refractory Ventricular Tachycardia to Drug and Ablation Therapy. J. Clin. Med. 2022, 11, 3549. https://doi.org/10.3390/jcm11123549

AMA Style

Shangguan W, Xu G, Wang X, Zhang N, Liu X, Li G, Tse G, Liu T. Stereotactic Radiotherapy: An Alternative Option for Refractory Ventricular Tachycardia to Drug and Ablation Therapy. Journal of Clinical Medicine. 2022; 11(12):3549. https://doi.org/10.3390/jcm11123549

Chicago/Turabian Style

Shangguan, Wenfeng, Gang Xu, Xin Wang, Nan Zhang, Xingpeng Liu, Guangping Li, Gary Tse, and Tong Liu. 2022. "Stereotactic Radiotherapy: An Alternative Option for Refractory Ventricular Tachycardia to Drug and Ablation Therapy" Journal of Clinical Medicine 11, no. 12: 3549. https://doi.org/10.3390/jcm11123549

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