Cardiac Sympathetic Denervation for Refractory Ventricular Arrhythmias

Marmar Vaseghi, Parag Barwad, Federico J Malavassi Corrales, Harikrishna Tandri, Nilesh Mathuria, Rushil Shah, Julie M Sorg, Jean Gima, Kaushik Mandal, Luis C Sàenz Morales, Yash Lokhandwala, Kalyanam Shivkumar, Marmar Vaseghi, Parag Barwad, Federico J Malavassi Corrales, Harikrishna Tandri, Nilesh Mathuria, Rushil Shah, Julie M Sorg, Jean Gima, Kaushik Mandal, Luis C Sàenz Morales, Yash Lokhandwala, Kalyanam Shivkumar

Abstract

Background: Cardiac sympathetic denervation (CSD) has been shown to reduce the burden of implantable cardioverter-defibrillator (ICD) shocks in small series of patients with structural heart disease (SHD) and recurrent ventricular tachyarrhythmias (VT).

Objectives: This study assessed the value of CSD and the characteristics associated with outcomes in this population.

Methods: Patients with SHD who underwent CSD for refractory VT or VT storm at 5 international centers were analyzed by the International Cardiac Sympathetic Denervation Collaborative Group. Kaplan-Meier analysis was used to estimate freedom from ICD shock, heart transplantation, and death. Cox proportional hazards models were used to analyze variables associated with ICD shock recurrence and mortality after CSD.

Results: Between 2009 and 2016, 121 patients (age 55 ± 13 years, 26% female, mean ejection fraction of 30 ± 13%) underwent left or bilateral CSD. One-year freedom from sustained VT/ICD shock and ICD shock, transplant, and death were 58% and 50%, respectively. CSD reduced the burden of ICD shocks from a mean of 18 ± 30 (median 10) in the year before study entry to 2.0 ± 4.3 (median 0) at a median follow-up of 1.1 years (p < 0.01). On multivariable analysis, pre-procedure New York Heart Association functional class III and IV heart failure and longer VT cycle lengths were associated with recurrent ICD shocks, whereas advanced New York Heart Association functional class, longer VT cycle lengths, and a left-sided-only procedure predicted the combined endpoint of sustained VT/ICD shock recurrence, death, and transplantation. Of the 120 patients taking antiarrhythmic medications before CSD, 39 (32%) no longer required them at follow-up.

Conclusions: CSD decreased sustained VT and ICD shock recurrence in patients with refractory VT. Characteristics independently associated with recurrence and mortality were advanced heart failure, VT cycle length, and a left-sided-only procedure.

Keywords: antiarrhythmic drugs; autonomic nervous system; functional class; implantable cardioverter-defibrillator; orthotopic heart transplantation.

Copyright © 2017 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.

Figures

FIGURE 1. Freedom from Individual Endpoints
FIGURE 1. Freedom from Individual Endpoints
Freedom from (A) sustained ventricular tachyarrhythmias (VT) or implantable cardioverter-defibrillator (ICD) shock as well as (B) freedom from death or orthoptopic heart transplantation (OHT) slowed after 1 year.
FIGURE 2. Sustained Survival
FIGURE 2. Sustained Survival
Similar sustained VT/ICD shock-free, transplant-free survival is seen in (A) the overall population and (B) in patients with ischemic cardiomyopathy (ICM) compared to patients with nonischemic cardiomyopathy (NICM). Abbreviations as in Figure 1.
FIGURE 3. Bilateral Versus Left CSD
FIGURE 3. Bilateral Versus Left CSD
There was a significant difference in the combined endpoint between bilateral and left cardiac sympathetic denervation (CSD). Abbreviations as in Figure 1.
FIGURE 4. Effect of NYHA Class on…
FIGURE 4. Effect of NYHA Class on Outcomes
Worsening New York Heart Association (NYHA) class significantly diminished CSD effectiveness on outcomes. Abbreviations as in Figures 1 and 3.
Figure 5. Pre-procedural Characteristics Associated with VT/ICD…
Figure 5. Pre-procedural Characteristics Associated with VT/ICD Shock Recurrence after CSD
Advanced NYHA class and longer VT cycle length (CL) predicted recurrence of sustained VT and ICD shock. AAD = antiarrhythmic drug; CI = confidence interval; CKD = chronic kidney disease; other abbreviations as in Figures 1, 3, and 4.
CENTRAL ILLUSTRATION. Pre-procedural Variables Associated With VT-free…
CENTRAL ILLUSTRATION. Pre-procedural Variables Associated With VT-free Transplant-free Survival after CSD
Patients with structural heart disease and recurrent ventricular tachyarrhythmias (VT) underwent cardiac sympathetic denervation (CSD) and were assessed for VT or implantable cardioverter-defibrillator (ICD) shock, death, or orthoptopic heart transplantation (OHT). On mulitvariable analysis, advanced New York Heart Association (NYHA) class, longer VT cycle lengths (CL), and a left-sided only procedure predicted the combined endpoint. AAD = antiarrhythmic drug; CI = confidence interval; CKD = chronic kidney disease; DMII = type 2 diabetes mellitus. Hazard ratio for VT CL refers to every 20 ms increase in cycle length.

References

    1. Vaseghi M, Shivkumar K. The role of the autonomic nervous system in sudden cardiac death. Prog Cardiovasc Dis. 2008;50:404–19.
    1. Zipes DP, Rubart M. Neural modulation of cardiac arrhythmias and sudden cardiac death. Heart Rhythm. 2006;3:108–13.
    1. Herrmann C, von zur Muhen F, Schaumann A, et al. Standardized assessment of psychological well-being and quality-of-life in patients with implanted defibrillators. Pacing Clin Electrophysiol. 1997;20:95–103.
    1. Poole JE, Johnson GW, Hellkamp AS, et al. Prognostic importance of defibrillator shocks in patients with heart failure. N Engl J Med. 2008;359:1009–17.
    1. Kumar S, Tedrow UB, Stevenson WG. Adjunctive Interventional Techniques When Percutaneous Catheter Ablation for Drug Refractory Ventricular Arrhythmias Fail: A Contemporary Review. Circ Arrhythm Electrophysiol. 2017;10:e003676.
    1. Shivkumar K, Ajijola OA, Anand I, et al. Clinical neurocardiology defining the value of neuroscience-based cardiovascular therapeutics. J Physiol. 2016;594:3911–54.
    1. Irie T, Yamakawa K, Hamon D, Nakamura K, Shivkumar K, Vaseghi M. Cardiac sympathetic innervation via middle cervical and stellate ganglia and antiarrhythmic mechanism of bilateral stellectomy. Am J Physiol Heart Circ Physiol. 2017;312:H392–H405.
    1. Schwartz PJ, Stone HL. Left stellectomy in the prevention of ventricular fibrillation caused by acute myocardial ischemia in conscious dogs with anterior myocardial infarction. Circulation. 1980;62:1256–65.
    1. Bourke T, Vaseghi M, Michowitz Y, et al. Neuraxial modulation for refractory ventricular arrhythmias: value of thoracic epidural anesthesia and surgical left cardiac sympathetic denervation. Circulation. 2010;121:2255–62.
    1. Schwartz PJ, Motolese M, Pollavini G, et al. Prevention of Sudden Cardiac Death After a First Myocardial Infarction by Pharmacologic or Surgical Antiadrenergic Interventions. J Cardiovasc Electrophysiol. 1992;3:2–16.
    1. Vaseghi M, Gima J, Kanaan C, et al. Cardiac sympathetic denervation in patients with refractory ventricular arrhythmias or electrical storm: intermediate and long-term follow-up. Heart Rhythm. 2014;11:360–6.
    1. Schoonmaker FW, Carey T, Grow JB., Sr Treatment of tachyarrhythmias and bradyarrhythmias by cardiac sympathectomy and permanent ventricular pacing. Ann Thorac Surg. 1975;19:80–7.
    1. Collura CA, Johnson JN, Moir C, Ackerman MJ. Left cardiac sympathetic denervation for the treatment of long QT syndrome and catecholaminergic polymorphic ventricular tachycardia using video-assisted thoracic surgery. Heart Rhythm. 2009;6:752–9.
    1. Marhold F, Izay B, Zacherl J, Tschabitscher M, Neumayer C. Thoracoscopic and anatomic landmarks of Kuntz’s nerve: implications for sympathetic surgery. Ann Thorac Surg. 2008;86:1653–8.
    1. Ramsaroop L, Partab P, Singh B, Satyapal KS. Thoracic origin of a sympathetic supply to the upper limb: the ‘nerve of Kuntz’ revisited. J Anat. 2001;199:675–82.
    1. Moss AJ, Schuger C, Beck CA, et al. Reduction in inappropriate therapy and mortality through ICD programming. N Engl J Med. 2012;367:2275–83.
    1. Hohnloser SH, Kuck KH, Dorian P, et al. Prophylactic use of an implantable cardioverter-defibrillator after acute myocardial infarction. N Engl J Med. 2004;351:2481–8.
    1. Tung R, Vaseghi M, Frankel DS, et al. Freedom from recurrent ventricular tachycardia after catheter ablation is associated with improved survival in patients with structural heart disease: An International VT Ablation Center Collaborative Group study. Heart Rhythm. 2015;12:1997–2007.
    1. Issa ZF, Zhou X, Ujhelyi MR, et al. Thoracic spinal cord stimulation reduces the risk of ischemic ventricular arrhythmias in a postinfarction heart failure canine model. Circulation. 2005;111:3217–20.
    1. Lopshire JC, Zhou X, Dusa C, et al. Spinal cord stimulation improves ventricular function and reduces ventricular arrhythmias in a canine postinfarction heart failure model. Circulation. 2009;120:286–94.
    1. Armaganijan LV, Staico R, Moreira DA, et al. 6-Month Outcomes in Patients With Implantable Cardioverter-Defibrillators Undergoing Renal Sympathetic Denervation for the Treatment of Refractory Ventricular Arrhythmias. JACC Cardiovasc Interv. 2015;8:984–90.
    1. Linz D, Wirth K, Ukena C, et al. Renal denervation suppresses ventricular arrhythmias during acute ventricular ischemia in pigs. Heart Rhythm. 2013;10:1525–30.
    1. Remo BF, Preminger M, Bradfield J, et al. Safety and efficacy of renal denervation as a novel treatment of ventricular tachycardia storm in patients with cardiomyopathy. Heart Rhythm. 2014;11:541–6.
    1. De Ferrari GM, Dusi V, Spazzolini C, et al. Clinical Management of Catecholaminergic Polymorphic Ventricular Tachycardia: The Role of Left Cardiac Sympathetic Denervation. Circulation. 2015;131:2185–93.
    1. Schwartz PJ, Priori SG, Cerrone M, et al. Left cardiac sympathetic denervation in the management of high-risk patients affected by the long-QT syndrome. Circulation. 2004;109:1826–33.
    1. Della Bella P, Baratto F, Tsiachris D, et al. Management of ventricular tachycardia in the setting of a dedicated unit for the treatment of complex ventricular arrhythmias: long-term outcome after ablation. Circulation. 2013;127:1359–68.
    1. Stevenson WG, Wilber DJ, Natale A, et al. Irrigated radiofrequency catheter ablation guided by electroanatomic mapping for recurrent ventricular tachycardia after myocardial infarction: the multicenter thermocool ventricular tachycardia ablation trial. Circulation. 2008;118:2773–82.
    1. Dinov B, Arya A, Bertagnolli L, et al. Early referral for ablation of scar-related ventricular tachycardia is associated with improved acute and long-term outcomes: results from the Heart Center of Leipzig ventricular tachycardia registry. Circ Arrhythm Electrophysiol. 2014;7:1144–51.
    1. Irie T, Yamakawa K, Hamon D, Nakamura K, Shivkumar K, Vaseghi M. Cardiac sympathetic innervation via the middle cervical and stellate ganglia and anti-arrhythmic mechanism of bilateral stellectomy. Am J Physiol Heart Circ Physiol. 2017;312:H392–405.
    1. Khalsa SS, Shahabi L, Ajijola OA, Bystritsky A, Naliboff BD, Shivkumar K. Synergistic application of cardiac sympathetic decentralization and comprehensive psychiatric treatment in the management of anxiety and electrical storm. Front Integr Neurosci. 2014;7:98.
    1. Malliani A, Lombardi F, Pagani M, Recordati G, Schwartz PJ. Spinal sympathetic reflexes in the cat and the pathogenesis of arterial hypertension. Clin Sci Mol Med Suppl. 1975;2:259s–60s.
    1. Malliani A, Recordati G, Schwartz PJ. Nervous activity of afferent cardiac sympathetic fibres with atrial and ventricular endings. J Physiol. 1973;229:457–69.
    1. Schwartz PJ, Foreman RD, Stone HL, Brown AM. Effect of dorsal root section on the arrhythmias associated with coronary occlusion. Am J Physiol. 1976;231:923–8.
    1. Vaseghi M, Lux RL, Mahajan A, Shivkumar K. Sympathetic stimulation increases dispersion of repolarization in humans with myocardial infarction. Am J Physiol Heart Circ Physiol. 2012;302:H1838–46.
    1. Opthof T, Coronel R, Vermeulen JT, Verberne HJ, van Capelle FJ, Janse MJ. Dispersion of refractoriness in normal and ischaemic canine ventricle: effects of sympathetic stimulation. Cardiovasc Res. 1993;27:1954–60.
    1. Opthof T, Misier AR, Coronel R, et al. Dispersion of refractoriness in canine ventricular myocardium. Effects of sympathetic stimulation. Circ Res. 1991;68:1204–15.
    1. Vaseghi M, Yamakawa K, Sinha A, et al. Modulation of regional dispersion of repolarization and T-peak to T-end interval by the right and left stellate ganglia. Am J Physiol Heart Circ Physiol. 2013;305:H1020–30.
    1. Yagishita D, Chui RW, Yamakawa K, et al. Sympathetic nerve stimulation, not circulating norepinephrine, modulates T-peak to T-end interval by increasing global dispersion of repolarization. Circ Arrhythm Electrophysiol. 2015;8:174–85.
    1. Ben-David J, Zipes DP. Differential response to right and left ansae subclaviae stimulation of early afterdepolarizations and ventricular tachycardia induced by cesium in dogs. Circulation. 1988;78:1241–50.
    1. Priori SG, Mantica M, Schwartz PJ. Delayed afterdepolarizations elicited in vivo by left stellate ganglion stimulation. Circulation. 1988;78:178–85.
    1. Opthof T, Dekker LR, Coronel R, Vermeulen JT, van Capelle FJ, Janse MJ. Interaction of sympathetic and parasympathetic nervous system on ventricular refractoriness assessed by local fibrillation intervals in the canine heart. Cardiovasc Res. 1993;27:753–9.
    1. Vaseghi M, Zhou W, Shi J, et al. Sympathetic innervation of the anterior left ventricular wall by the right and left stellate ganglia. Heart Rhythm. 2012;9:1303–9.
    1. Janes RD, Brandys JC, Hopkins DA, Johnstone DE, Murphy DA, Armour JA. Anatomy of human extrinsic cardiac nerves and ganglia. Am J Cardiol. 1986;57:299–309.
    1. Kawashima T. Anatomy of the cardiac nervous system with clinical and comparative morphological implications. Anat Sci Int. 2011;86:30–49.
    1. Ajijola OA, Yagishita D, Reddy NK, et al. Remodeling of stellate ganglion neurons after spatially targeted myocardial infarction: Neuropeptide and morphologic changes. Heart Rhythm. 2015;12:1027–35.
    1. Armour JA. Activity of in situ middle cervical ganglion neurons in dogs, using extracellular recording techniques. Can J Physiol Pharmacol. 1985;63:704–16.

Source: PubMed

3
Abonnieren