ArticlePDF Available

Vagus nerve stimulation: An evolving adjunctive treatment for cardiac disease

Authors:

Abstract and Figures

The vagus nerve is a major component of the autonomic nervous system and plays a critical role in many body functions including for example, speech, swallowing, heart rate and respiratory control, gastric secretion, and intestinal motility. Vagus nerve stimulation (VNS) refers to any technique that stimulates the vagus nerve, with electrical stimulation being the most important. Implantable devices for VNS are approved therapy for refractory epilepsy and for treatment-resistant depression. In the case of heart disease applications, implantable VNS has been shown to be beneficial for treating heart failure in both preclinical and clinical studies. Adverse effects of implantable VNS therapy systems are generally associated with the implantation procedure or continuous on-off stimulation. The most serious implantation-associated adverse effect is infection. The effectiveness of non-invasive transcutaneous VNS for epilepsy, depression, primary headaches, heart failure, and other conditions remains under investigation. VNS merits further study for its potentially favorable effects on cardiovascular disease, especially heart failure.
Content may be subject to copyright.
Address for correspondence: Dr. Barış Akdemir, Mail Code 508, 420 Delaware St SE
Minneapolis, MN, 55455-USA
E-mail: akdem002@umn.edu
Accepted Date: 10.06.2016
©Copyright 2016 by Turkish Society of Cardiology - Available online at www.anatoljcardiol.com
DOI:10.14744/AnatolJCardiol.2016.7129
804 Review
Barış Akdemir, David G. Benditt
Cardiovascular Division, Cardiac Arrhythmia and Syncope Center, University of Minnesota Medical School; Minneapolis, Minnesota-
USA
Vagus nerve stimulation: An evolving adjunctive treatment
for cardiac disease
Introduction
The sympathetic and parasympathetic components of the au-
tonomic nervous system (ANS) regulate the physiological func-
tion of a wide range of organs, glands, and involuntary muscles;
conversely, the ANS may also contribute importantly to both the
development and treatment of disease process in these very
same organ systems. By way of example, in cardiovascular medi-
cine, autonomic neural inuences play a crucial role in determin-
ing the clinical features and severity of a wide range of condi-
tions including hypertension, ischemic arrhythmia, heart failure,
and reex syncope (1). Further, drugs with predominant impact
on autonomic function (e.g., beta- and alpha-adrenergic blockers,
most antiarrhythmic agents, and angiotensin receptor blockers)
are the foundation for treatment of many of these abnormalities.
Additionally, apart from drugs, recently there has been increased
interest in electrical ANS stimulation for treatment of certain of
these disease states. In this regard, the role of direct neural stimu-
lation for therapeutic application may be dated to initial attempts
to stimulate the carotid sinus for amelioration of severe angina
pectoris (1, 2). However, as more effective medical and surgical
techniques were introduced, the carotid sinus electrical stimula-
tion approach largely vanished. On the other hand, indirect elec-
trical stimulation of the heart, and inevitably its peripheral nerves,
has been the subject of a number of clinical trials targeting treat-
ment of certain reex syncopal disorders, particularly carotid
sinus syndrome and vasovagal syncope (1, 3). While these latter
clinical trials have met with variable success, they have spurred a
resurgence of research designed to identify the potential clinical
utility of modifying ANS activity by direct electrical stimulation.
Perhaps the ANS region that offers the greatest current inter-
est for direct electrical stimulation is that of the complex neural
networks residing on the posterior aspect of the heart (particu-
larly the atria) (1, 4). These neural complexes communicate with
the central nervous system via neural connections traveling
predominantly along the great vessels of the thorax. In a recent
review we summarized the body of research examining these
complex networks and their probable contributions to cardiac
arrhythmias, including potentially life-threatening channelopa-
thies (4). In terms of current therapeutics, stimulation of certain
aspects of these neural networks, particularly the regions adja-
cent to the pulmonary veins, plays a role in certain atrial bril-
lation ablation strategies. In essence, induction of bradycardia
by atrial stimulation in the vicinity of the neural network of inter-
The vagus nerve is a major component of the autonomic nervous system and plays a critical role in many body functions including for example,
speech, swallowing, heart rate and respiratory control, gastric secretion, and intestinal motility. Vagus nerve stimulation (VNS) refers to any
technique that stimulates the vagus nerve, with electrical stimulation being the most important. Implantable devices for VNS are approved
therapy for refractory epilepsy and for treatment-resistant depression. In the case of heart disease applications, implantable VNS has been
shown to be beneficial for treating heart failure in both preclinical and clinical studies. Adverse effects of implantable VNS therapy systems
are generally associated with the implantation procedure or continuous on-off stimulation. The most serious implantation-associated adverse
effect is infection. The effectiveness of non-invasive transcutaneous VNS for epilepsy, depression, primary headaches, heart failure, and other
conditions remains under investigation. VNS merits further study for its potentially favorable effects on cardiovascular disease, especially heart
failure. (Anatol J Cardiol 2016; 16: 804-10)
Keywords: atrial fibrillation, heart failure, vagus nerve stimulation, ventricular arrhythmia
ABSTRACT
est is used to conrm proximity for purposes of radio-frequency
modication of efferent ANS inputs to the heart, which most like-
ly also alter afferent signals to the mid-brain (5, 6). Although the
ganglionic ablation strategy for atrial brillation ablation remains
controversial, the concept of direct ANS stimulation to identify a
ganglionic target continues to be employed in many clinical elec-
trophysiological laboratories for difcult cases (6).
Apart from targeting ganglionic plexus (GP), there are vari-
ous elements of the ANS that may be amenable to functional
modication by direct electrical stimulation. In this context, the
vagus nerve (10th cranial nerve) has the virtue of being readily
accessible (vagus nerve stimulation, VNS). The vagus is princi-
pally a mixed parasympathetic nerve, containing both afferent
and efferent sensory bers. Vagal nerve activity plays a promi-
nent role in heart rate and respiratory control, gastric secretion,
and intestinal motility. In addition, vagus nerve connections mod-
ulate the function of higher brain centers, forming the basis for
its potential use in many clinical disorders (1, 3, 4). For instance,
the vagus nerve plays a key role in blood pressure (BP) control.
Further, in conjunction with sympathetic “withdrawal” in reex
vasodepressor syncope, increased parasympathetic activity
largely traveling in the vagus nerve can act to substantially re-
duce BP. This latter attribute was investigated in our laboratory
and we demonstrated that enhanced vagal activity triggered in-
directly by carotid sinus stimulation acted to reduce systemic
BP even in the setting of sympathetic blockade and absence of
cardiac slowing (7). This and other similar observations provide
a reasonable basis for assessing the potential for direct vagal
stimulation to contribute to BP control in difcult to treat pa-
tients, and by virtue of afterload reduction, to possibly play a role
in treatment of both low cardiac output states, and diminishing
arrhythmia susceptibility in systolic heart failure (8).
Apart from its potential value for cardiovascular disease,
electrical stimulation of the vagus (either directly or indirectly)
has proved useful in treatment of a number of other medical
conditions. In this regard, electrical VNS has US Food and Drug
Administration (FDA) approval for management of epilepsy and
depression. In addition, VNS is being studied for possible ben-
ets in headache, gastric motility disorders, and asthma (9). This
article focuses primarily on development of clinical VNS for car-
diac applications, including consideration of VNS device types
(invasive or noninvasive), and potential adverse effects.
Development of clinical VNS
VNS and epilepsy
In the late 19th century, VNS was rst used to treat epilepsy
by American neurologist James Corning, but the method was as-
sociated with excessive adverse effects (e.g., bradycardia, syn-
cope) and was abandoned (10). More recently the concept has
been resurrected and is used clinically.
VNS effectiveness in epilepsy was demonstrated with early
animal studies (11, 12). Subsequently, clinical studies of implant-
able VNS Therapy System® (Cyberonics, Inc., Houston, TX, USA,
Fig.1) in patients with refractory epilepsy, showed a seizure
reduction of ≥50% in 24.5% to 46.6% of patients (13). The VNS
Therapy System® was approved for the treatment of medically
refractory epilepsy in Europe in 1994 and in the USA and Canada
in 1997. As of August 2014, over 100,000 VNS devices had been
implanted in more than 75,000 patients worldwide (14).
The mechanism(s) of VNS benet for epilepsy prevention
remains largely unknown. However, in this regard, we have re-
cently reported that ictal asystole may be a model for improving
understanding of 1 set of cortical sites that may trigger both va-
gal bradycardia and vasodepression mimicking a reex faint. In
essence, focal epilepsy arising in the right or left insular cortex
has been associated with both a drop in BP and at times brady-
cardia, and thereby may reect 1 cortical region in which electri-
cal stimulation may modify susceptibility to epilepsy, as well as
benecially reduce BP when desired (15, 16).
VNS and depression
VNS has also found a role in the management of treatment-
resistant depression (TRD). Several observations led to consid-
eration of this application, including in particular improvement
of mood and cognition in epilepsy patients after initiation of VNS
therapy, and usage of several anticonvulsant medications as
mood stabilizers and antidepressants in bipolar disorder.
Brain regions that are critical to mood regulation (orbital cor-
tex, limbic system) are targets for VNS. Rush et al. (17) designed
a study to investigate effect of VNS on TRD. In short-term VNS
therapy for TRD, there was no statistical difference between
VNS therapy “on” versus VNS “off,” in terms of the 24-item
Hamilton Depression Rating Scale (HRSD24) response. However,
the study was extended to 1 year in 205 patients, and ndings
indicated that the HRSD24 score improved signicantly in VNS
therapy group (p<0.001) (18). Based on these observations, VNS
therapy was approved by FDA for TRD patients ≥18 years old (19).
VNS and heart disease
As noted earlier, neural stimulation for amelioration of car-
diovascular disease has been the subject of study for many
Akdemir et al.
Vagus nerve stimulationAnatol J Cardiol 2016; 16: 804-10
Figure 1. Implantable vagus nerve stimulation therapy system (Car-
diotTM, BioControl-Medical, Yehud, Israel; Pulse Model 102 Genera-
tor, Cyberonics, Inc., Houston, TX, USA,)
805
years. Early interest focused on carotid sinus stimulation for
intractable angina and later for treatment of hypertension (2).
These applications are summarized further below, but were
based on the already well-known propensity for carotid sinus
massage to decrease heart rate and BP. More recently, direct
VNS has begun to be of special interest as an adjuvant therapy
in heart failure patients.
Carotid sinus nerve stimulation (CsNS)
for angina pectoris and hypertension
Stimulation of baroreceptors in the carotid sinuses and aortic
arch results in reex systemic arterial and splanchnic bed dila-
tion, and reduction of both heart rate and myocardial contracti-
lity (20). The heart rate and contractility changes occur as a con-
sequence of reduction in the frequency of sympathetic efferent
impulses to sinus node and ventricular muscle, and an increase
in the frequency of vagal impulses (21).
In the mid-20th century CsNS gained interest as a potential
means for alleviating drug-refractory angina pectoris, by de-
creasing myocardial oxygen consumption (i.e., decreased heart
rate and contractility) at a time when it was not possible to im-
prove myocardial blood supply (1, 22). Braunwald et al. (23), in
a landmark report, showed that carotid nerve stimulation de-
creased angina episodes and increased exercise tolerance in 15
of 22 patients who had stable coronary artery disease. However,
CsNS never became a mainstream therapy for angina pectoris
due to advances in both pharmacological and coronary reperfu-
sion strategies.
CsNS has also been investigated for systemic hypertension
for more than 40 years. An implantable CsNS therapy system
(Barostim neo System, CVRx Inc., Minneapolis, MN, USA, Fig. 2)
has CE (Conformité Européene) mark in Europe for the treatment
of hypertension patients. This device is currently under clinical
evaluation in the USA and Canada for the treatment of high blood
pressure and heart failure (24).
VNS for heart failure
That autonomic disturbances contribute importantly to the
progression of heart failure is now widely recognized (1, 22). In
this context, autonomic imbalance characterized by vagal with-
drawal and increased sympathetic activity has been shown to
play a major role in the worsening of both heart failure and its
prognosis. Specically, while sympathetic pre-dominance may
be benecial in acute cardiac events to maintain cardiac output,
chronically excessive sympathetic activity is detrimental, con-
tributing to adverse cellular calcium loading, left ventricular (LV)
remodeling, myocyte apoptosis, brosis, and electrical instability.
Clinical evidence from the Autonomic Tone and Reexes
after Myocardial Infarction study (25) and the Cardiac Insuf-
ciency Bisoprolol Study II (26) indicates that diminished cardiac
vagal activity and increased heart rate predict a high mortality
rate in congestive heart failure. Therefore, modulation of the
ANS (neuromodulation) with the aim of restoring a more normal
autonomic balance is gaining increasing interest as a potential
therapy for patients with heart failure. In this regard, it is hypoth-
esized that electrical stimulation of the vagus nerve may help to
normalize parasympathetic activation of cardiac control reex-
es and inhibit sympathetic hyperactivation (1, 27).
In preclinical studies, VNS has demonstrated improved
cardiac electrical and mechanical function. For instance, Li et
al. (28) showed improvement in hemodynamics, LV remodeling
and reduced neurohormonal activation with VNS in a rat infarct
model with heart failure. Study results showed a reduction in
mortality rate at 140 days (50% in the sham model and 14% with
VNS stimulation) (28).
An initial clinical study for heart failure included patients
with New York Heart Association (NYHA) class II-III heart failure
and LV ejection fraction (LVEF) <35%. This report demonstrated
improvement of LV end-systolic volume (LVESV), NYHA classi-
cation, and quality of life (29). A subsequent report on patients
with reduced EF and NYHA classes II-IV showed improvement in
LVEF, cardiac volume, and 6-minute walk test at 6 months, which
was maintained at 1 year (30).
The Increase of Vagal Tone in Heart Failure (INOVATE-HF)
trial (CardiotTM VNS therapy system; BioControl Medical, Ye-
hud, Israel, Fig. 1) is similarly assessing VNS in heart failure and
has just recently achieved target of 650 patients. Findings are
expected in December 2016.
Neural Cardiac Therapy for Heart Failure (Precision, Bos-
ton Scientic Corporation, St. Paul, MN, USA) was a random-
ized controlled trial of VNS in patients with EF <35%, increased
LV end-diastolic dimensions (>55 mm), and NYHA classes III-IV,
but excluding patients with cardiac resynchronization therapy
devices; or QRS>130 milliseconds (31). Patients were random-
ized in a 2:1 fashion to VNS on or off for 6 months. The primary
endpoint was improvement in LV systolic dimensions; secondary
endpoints were improvement in other echocardiographic param-
Akdemir et al.
Vagus nerve stimulation Anatol J Cardiol 2016; 16: 804-10
Figure 2. Carotid sinus nerve stimulation therapy system (Barostim
neo System, CVRx Inc., Minneapolis, MN, USA)
806
eters and circulating biomarkers. The study failed to reach pri-
mary or secondary endpoints; however, it did show improvement
in quality of life and NYHA classication (31).
The Autonomic Neural Regulation Therapy to Enhance Myo-
cardial Function in Heart Failure (Cyberonics, Houston, TX, USA)
study investigated VNS of right or left cervical vagus in 60 patients
(32). The main inclusion criteria were EF <40%, LV end-diastolic
dimensions 50–80 mm, and QRS<150 milliseconds. There was
improvement in LVEF by 4.5%, but LVESV did not decrease signi-
cantly. There was again an improvement in quality of life, exercise
capacity, and NYHA classication. There was no signicant dif-
ference between left or right cervical vagus stimulation (32).
VNS for cardiac arrhythmias
Atrial brillation
As discussed earlier, supra-threshold VNS (i.e., electrical
stimulation of the vagus nerve at a voltage level that slows the
sinus rate or prolongs atrioventricular conduction) has been stu-
died in other cardiac and non-cardiac diseases. Supra-thresh-
old VNS has been used to induce and maintain atrial brillation
(AF), and animal studies have shown that supra-threshold VNS
can be utilized to induce and maintain AF in experiments. How-
ever, recent experimental and clinical studies show that low-lev-
el cervical VNS (i.e., the voltages/currents do not slow the sinus
rate or prolong atrioventricular conduction) induces effects op-
posite to supra-threshold VNS and plays an anti-arrhythmic role
in AF management. Since 2009, several studies have reported
the anti-arrhythmic role of low-level cervical VNS in AF popu-
lation (33–36). For example, Li et al. (33) showed that cervical
low-level VNS induced a progressive increase in AF threshold at
all pulmonary vein and atrial appendage sites. Yu et al. (34) also
demonstrated that cervical low-level VNS inhibited AF inducibil-
ity, prevented shortening of effective refractory period (ERP) at
pulmonary vein and atrial sites and increase of ERP dispersion
induced by activation of atrial GP. Finally, a series of recent stud-
ies also showed that cervical low-level VNS can prevent and/
or reverse atrial electrophysiological remodeling and autonomic
remodeling (35, 36).
Cervical VNS is an invasive approach in which cervical sur-
gery is needed to position vagal stimulation electrode and has
adverse effects that are discussed in detail below. Recently, a
noninvasive approach for VNS (nVNS), transcutaneous electri-
cal stimulation of the auricular branch of the vagus nerve located
at the tragus, has been used in some studies (Fig. 3). In these
studies, it has been shown that low-level nVNS suppressed
AF, reversed acute atrial electrophysiological remodeling, de-
creased sympathetic nerve activity and increased heart rate
variability (37, 38).
Ventricular arrhythmias
As noted earlier, different levels of parasympathetic stimu-
lation exert different anti-arrhythmic effects. Thus, it has been
shown that parasympathetic hyperactivity induces and main-
tains AF, but at the same time, parasympathetic hyperactivity
is protective for ventricular arrhythmias. In animal studies, cer-
vical supra-threshold VNS suppressed the incidence of ven-
tricular arrhythmias, especially ventricular tachycardia and
ventricular brillation during acute myocardial ischemia and
ischemia reperfusion (39, 40). Multiple mechanisms have been
described to explain this anti-arrhythmic effect of VNS, such as
its bradycardiac effect, anti-adrenergic effects, prevention of
the loss of phosphorylated connexin 43 proteins, and inhibition
of the opening of the mitochondrial permeability transition pore.
The anti-arrhythmic role of atrial GP stimulation has also been
investigated in several studies. In the normal heart, activation of
atrial GP prolonged ventricular ERP, decreased the slope of vent-
ricular action potential restitution curves and delayed action
potential duration (41). In the ischemic heart, atrial GP activity
signicantly inhibited the incidence of ventricular arrhythmias
during not only acute myocardial ischemia (42), but also isch-
emia reperfusion (43). Atrial GP stimulation also increased heart
rate variability and prevented the loss of connexin 43 induced by
ischemia/reperfusion (43).
VNS device types and adverse effects
Implantable VNS appears to be safe and well tolerated.
The electrodes are attached to the left or right vagus nerve
connected to a stimulating device implanted under the ante-
rior chest wall (Fig. 1). Stimulation is turned on or off by a mag-
net. The VNS device may operate using a wide variety of stim-
ulation parameters (output current, signal frequency, pulse
width, signal on time, signal off time). Currently approved
stimulation parameters are 0.25–3.5 mA (0.25 mA steps), 20–30
Hz (<10 Hz is ineffective, >50 Hz might induce nerve damage),
0.25–0.5 milliseconds pulse, signal on for 30–60 seconds, and
signal off for 5 minutes. (44, 45) A rapid stimulation of signal
on for 7 seconds and off for 14-21 seconds is also available
(46). The optimal VNS stimulation settings, however, remain
unknown.
Figure 3. Non-implantable vagus nerve stimulation systems: (a)
NEMOS® (transcutaneous vagus nerve stimulation; Cerbomed, Erlan-
gen, Germany), (b) gammaCore (noninvasive vagus nerve stimulation;
ElectroCore, Basking Ridge, NJ, USA)
a b
Akdemir et al.
Vagus nerve stimulationAnatol J Cardiol 2016; 16: 804-10 807
Adverse events (AEs) with VNS are generally associated
with implantation or continuous on-off stimulation. As is the
case with any implanted device, infection is the most serious
implantation-associated complication. Bradycardia and asys-
tole have also been described during implantation, as has vocal
cord palsy, which has been noted to persist up to 6 months, and
occurrence depends on surgical skill and experience. Recently,
a retrospective study (47) was published that was designed to
investigate surgical and hardware-related complications of VNS
implantation. Complications related to surgery occurred in 8.6%
and hardware complications in 3.7%. Table 1 summarizes comp-
lication rates related to surgery and hardware. Complication
rates in the rst 10 years of implantation experience were com-
pared with last 15 years implantation experience; similar surgi-
cal complication rates were found in both groups (8.9%, 9.2%)
but hardware-related complication rates were less in the last 15
years experience (7.3%, 2.3%).
The most frequent stimulation-associated AEs include voice
alteration, paresthesia, cough, headache, dyspnea, pharyngi-
tis, and pain. Table 2 summarizes stimulation-associated AEs in
clinical trials of the VNS Therapy System. The frequency of these
AEs declines with continued treatment (48). Treatment will likely
require a decrease in stimulation strength or intermittent or per-
manent device deactivation (9).
Alternative nVNS delivery systems such as that stimulating
the tragus of the ear avoid surgery-related complications and may
limit AEs related to the continuous on-off stimulation cycle of im-
plantable devices since nVNS devices can be adjusted to balance
efcacy and tolerability (49, 50). Efcacy could not be compared
between implantable VNS system and nVNS system at the time of
this review. Randomized prospective studies are needed to com-
pare efcacy of nVNS with implantable VNS therapy system.
Summary
Neuromodulation offers a potentially important new ap-
proach to enhance treatment of a range of cardiovascular disea-
ses. VNS is currently the neuromodulation method that has so far
received most clinical interest. Specically, by modifying sympa-
thetic/parasympathetic balance to the heart and other cardio-
vascular structures, VNS may offer an adjunct to conventional
treatment strategies for a number of inadequately controlled
cardiovascular conditions. At present, heart failure provides
the most important potential application, but additional study is
needed to ascertain whether VNS will provide substantial incre-
mental benet. However, other possible VNS opportunities may
include ameliorating inappropriate sinus tachycardia, preventing
AF and reducing propensity for sudden death in certain high-risk
populations.
Potential conflicts of interest: Dr. Benditt was an investigator for
the INOVATE-HF trial, but otherwise has no commercial or nancial con-
icts to declare. Dr. Benditt is supported in part by a grant from the Earl
E. Bakken family in support of heart-brain research.
Peer-review: Externally peer-reviewed.
Authorship contributions: Concept – D.B.; Design – D.B.; Supervi-
sion – D.B.; Literature search – B.A.; Writing – B.A.; Critical review – D.B.
References
1. Benditt DG, Iskos D, Sakaguchi S. Autonomic nervous system and
cardiac arrhythmias. In. Electrophysiological Discorders of the
Heart. (eds. Sakasena S and Camm AJ) Philedelphia, PA, Elsevier
Chrchill Livingstone 2005.p. 49-67. Crossref
2. Ermiş C, Benditt DG. Carotid sinus massage during evaluation for
transient loss of consciousness: just a positive test. Europace 2004;
6: 292-5. Crossref
3. Benditt DG, Lurie KG, Fahy G, Iskos D, Sakaguchi S. Treatment of
vasovagal syncope: Is there a role for cardiac pacing? In. Non-
pharmacological Therapy of Arrhythmias for the 21st Centrury: The
State of the Art (eds. Singer I, Barold SS, Camm AJ) Armonk, NY,
Futura Publishing Co, Inc., 1998.p. 881-9.
4. Benditt DG, Sakaguchi S, van Dijk JG. Autonomic Nervous System
and Cardiac Arrhythmias. In. Electrophysiology Disorders of the
Heart 2nd edition. Saksena S, Camm AJ, Boyden PA, Dorian P, Gold-
schlager N, Vetter , Zareba W (eds). Elsevier Sauders, Philadelphia,
2012. p. 61-71. Crossref
5. Benditt DG, Samniah N, Fahy GJ, Lurie KG, Sakaguchi S. Atrial Fi-
brillation: defining potential curative ablation targets. J Interv Card
Table 1. Complications of implantable VNS device related to surgery
and hardware (47)
Complications related to surgery Rate (%)
Hematoma 1.9
Infection 2.6
Vocal cord palsy 1.4
Lower facial weakness <1
Pain and sensory-related complications 1.4
Bradycardia <1
Complications related to hardware Rate (%)
Lead fracture/lead malfunction 3
Spontaneous VNS turn-on <1
Lead disconnection <1
VNS - vagus nerve stimulation
Table 2. Stimulation-associated adverse events in clinical trials of the
VNS Therapy System (48)
Adverse event 3 months (%) 12 months (%) 5 year (%)
Cough 21 15 1.5
Voice alteration 62 55 18.7
Dyspnea 16 13 2.3
Pain 17 15 4.7
Paresthesia 25 15 1.5
Headache 20 16 NA
VNS: vagus nerve stimulation
Akdemir et al.
Vagus nerve stimulation Anatol J Cardiol 2016; 16: 804-10
808
Electrophysiol 2000; 4: 141-7. Crossref
6. Lu F, Adkisson WO, Chen T, Akdemir B, Benditt DG. Catheter abla-
tion for long-standing persistent atrial fibrillation in patients who
have failed electrical cardioversion. J Cardiovasc Transl Res 2013;
6: 278-86. Crossref
7. Almquist A, Gornick C, Benson WJr, Dunnigan A, Benditt DG. Ca-
rotid sinus hypersensitivity: evaluation of the vasodepressor com-
ponent. Circulation 1985; 71: 927-36. Crossref
8. Reiter MJ, Stromberg KD, Whitman TA, Adamson PB, Benditt DG,
Gold MR. Influence of intra-cardiac pressure on spontaneous ven-
tricular arrhythmias in patients with systolic heart failure: Insights
from the REDUCEhf trial. Circ Arrhythm Electrophysiol 2013; 6: 272-8.
9. Ben-Menachem E, Revesz D, Simon BJ, Silberstein S. Surgically
implanted and non-invasive vagus nerve stimulation: a review of
efficacy, safety and tolerability. Eur J Neurol 2015; 22: 1260-8.
10. Lanska JL. Corning and vagal nerve stimulation for seizures in the
1880s. Neurology 2002; 58: 452-9. Crossref
11. Lockard JS, Congdon WC, DuCharme LL. Feasibility and safety of
vagal stimulation in monkey model. Epilepsia 1990; 31 (Suppl. 2):
S20-6. Crossref
12. Woodbury JW, Woodbury DM. Vagal stimulation reduces the sever-
ity of maximal electroshock seizures in intact rats: use of a cuff
electrode for stimulating and recording. Pacing Clin Electrophysiol
1991; 14: 94-107. Crossref
13. Ben-Menachem E, Manon-Espaillat R, Ristanovic R, Wilder BJ,
Stefan H, Mirza W, et al. Vagus nerve stimulation for treatment of
partial seizures: 1. A controlled study of effect on seizures. First
International Vagus Nerve Stimulation Study Group. Epilepsia 1994;
35: 616-26. Crossref
14. Cyberonics Inc. 2013 Annual Report. http://ir.cyberonics.com/annu-
als.cfm.
15. Kohno R, Abe H, Akamatsu N, Oginosawa Y, Tamura M, Takeuchi M,
et al. Syncope and ictal asystole caused by temporal lobe epilepsy.
Circ J 2011; 75: 2508-10. Crossref
16. Benditt DG, van Dijk G, Thijs RD. Ictal asystole: life-threatening va-
gal storm or a benign seizure self-termination mechanism? Circ Ar-
rhythmi Electrophysiol 2015; 8: 11-4. Crossref
17. Rush AJ, Marangell LB, Sackeim HA, George MS, Brannan SK,
Davis SM, et al. Vagus nerve stimulation for treatment-resistant
depression: a randomized, controlled acute phase trial. Biol Psy-
chiatry 2005; 58: 347-54. Crossref
18. Rush AJ, Sackeim HA, Marangell LB, George MS, Brannan SK, Da-
vis SM, et al. Effects of 12 months of vagus nerve stimulation in
treatment-resistant depression: a naturalistic study. Biol Psychia-
try 2005; 58: 355-63. Crossref
19. VNS Therapy System Physician’s Manual. Houston, TX: Cyberonics
Inc., 2013. http://dynamic.cyberonics.com/manuals.
20. Samniah N, Sakaguchi S, Ermis C, Lurie KG, Benditt DG. Transient
modification of baroreceptor response during tilt-induced vasova-
gal syncope. Europace 2004; 6: 48-54. Crossref
21. Kezdi P. Baroreceptors and Hypertension. New York Pergamon
Press, 1967.
22. Sutton R, Fisher JD, Linde C, Benditt DG. History of electrical thera-
py for the heart. Eur Heart J (Suppl) 2007; Suppl I: 13-110.
23. Braunwald E, Epstein SE, Glick G, Wechsler AS, Braunwald NS. Re-
lief of angina pectoris by electrical stimulation of the carotid sinus
nerves. N Engl J Med 1967; 277: 1278-83. Crossref
24. CVRx Inc. Clinical evidence. www.cvrx.com/clinical evidence.
25. La Rovere MT, Bigger JT Jr, Marcus FI, Mortara A, Schwartz PJ.
Baroreflex sensitivity and heart-rate variability in prediction of total
cardiac mortality after myocardial infarction. Lancet 1998; 351: 478-
84. Crossref
26. Lechat P, Hulot JS, Escolano S, Mallet A, Leizorovicz A, Werhlen-
Grandjean M, et al. Heart rate and cardiac rhythm relationships
with bisoprolol benefit in chronic heart failure in CIBIS II trial. Cir-
culation 2001; 103: 1428-33. Crossref
27. Sutton R, Brignole M, Benditt DG. Key challenges in the current
management of syncope. Nat Rev Cardiol 2012; 9: 590-8. Crossref
28. Li M, Zheng C, Sato T, Kawada T, Sugimachi M, Sunagawa K. Va-
gal nerve stimulation markedly improves long-term survival after
chronic heart failure in rats. Circulation 2004; 109: 120-4. Crossref
29. Schwartz PJ, De Ferrari GM, Sanzo A, Landolina M, Rordorf R, Rai-
neri C, et al. Long term vagal stimulation in patients with advanced
heart failure: first experience in man. Eur J Heart Fail 2008; 10: 884-
91. Crossref
30. De Ferrari GM, Crijns HJ, Borggrefe M, Milasinovic G, Smid J, Zabel
M, et al. CardioFit Multicenter Trial Investigators. Chronic vagus
nerve stimulation: a new and promising therapeutic approach for
chronic heart failure. Eur Heart J 2011; 32: 847-55. Crossref
31. Zannad F, De Ferrari GM, Tuinenburg AE, Wright D, Brugada J, But-
ter C, et al. Chronic vagal stimulation for the treatment of low ejec-
tion fraction heart failure: results of the NEural Cardiac TherApy
foR Heart Failure (NECTAR-HF) randomized controlled trial. Eur
Heart J 2015; 36: 425-33. Crossref
32. Premchand RK, Sharma K, Mittal S, Monteiro R, Dixit S, Libbus I,
et al. Autonomic regulation therapy via left or right cervical vagus
nerve stimulation in patients with chronic heart failure: results of
the ANTHEM-HF Trial. J Card Fail 2014; 20: 808-16. Crossref
33. Li S, Scherlag BJ, Yu L, Sheng X, Zhang Y, Ali R, et al. Low-level
vagosympathetic stimulation:a paradox and potential new modal-
ity for the treatment of focal atrial fibrillation. Circ Arrhythm Elec-
trophysiol 2009; 2: 645-51. Crossref
34. Yu L, Scherlag BJ, Li S, Sheng X, Lu Z, Nakagawa H, et al. Low-level
vagosympathetic nerve stimulation inhibits atrial fibrillation induc-
ibility: direct evidence by neural recordings from intrinsic cardiac
ganglia. J Cardiovasc Electrophysiol 2011; 22: 455-63. Crossref
35. Sheng X, Scherlag BJ, Yu L, Li S, Ali R, Zhang Y, et al. Prevention and
reversal of atrial fibrillation inducibility and autonomic remodeling
by low-level vagosympathetic nerve stimulation. J Am Coll Cardiol
2011; 57: 563-71. Crossref
36. Sha Y, Scherlag BJ, Yu L, Sheng X, Jackman WM, Lazzara R, et al.
Low-level right vagal stimulation: anticholinergic and antiadrener-
gic effects. J Cardiovasc Electrophysiol 2011; 22: 1147-53. Crossref
37. Yu L, Scherlag BJ, Li S, Fan Y, Dyer J, Male S, et al. Low-level trans-
cutaneous electrical stimulation of the auricular branch of the va-
gus nerve: A noninvasive approach to treat the initial phase of atrial
fibrillation. Heart Rhythm 2013; 10: 428-35. Crossref
38. Clancy JA, Mary DA, Witte KK, Greenwood JP, Deuchars SA, Deuc-
hars J. Non-invasive vagus nerve stimulation in healthy humans
reduces sympathetic nerve activity. Brain Stimul 2014; 7: 871-7.
39. Myers RW, Pearlman AS, Hyman RM, Goldstein RA, Kent KM, Gold-
stein RE, et al. Beneficial effects of vagal stimulation and brady-
cardia during experimental acute myocardial ischemia. Circulation
1973; 49: 943-7. Crossref
40. Zuanetti G, De Ferrari GM, Priori SG, Schwartz PJ. Protective effect
of vagal stimulation on reperfusion arrhythmias in cats. Circ Res
1987; 61: 429-35. Crossref
41. He B, Lu Z, He W, Huang B, Yu L, Wu L, et al. The effects of atrial
ganglionated plexi stimulation on ventricular electrophysiology in a
normal canine heart. J Interv Card Electrophysiol 2013; 37: 1-8.
Akdemir et al.
Vagus nerve stimulationAnatol J Cardiol 2016; 16: 804-10 809
42. He B, Lu Z, He W, Wu L, Huang B, Yu L, et al. Effects of low-in-
tensity atrial ganglionated plexi stimulation on ventricular elec-
trophysiology and arrhythmogenesis. Auton Neurosci 2013; 174:
54-60. Crossref
43. Yu L, He W, Huang B, He B, Jiang H. Atrial ganglionated plexus
stimulation prevents myocardial ischemia reperfusion arrhythmias
by preserving connexin43 protein. Circulation 2012; 126: A18522.
44. Agnew WF, McCreery DB. Considerations for safety with chroni-
cally implanted nerve electrodes. Epilepsia 1990; 31 Suppl 2:S27-
S32. Crossref
45. Terry Jr RS. Vagus nerve stimulation therapy for epilepsy. In: Holmes
M, ed. Epilepsy Topics: InTech; 2014. p.139-60. Crossref
46. Morris GL 3rd, Gloss D, Buchhalter J, Mack KJ, Nickels K, Harden
C. Evidence-based guideline update: Vagus nerve stimulation for
the treatment of epilepsy: report of the Guideline Development
Subcommittee of the American Academy of Neurology.Neurology
2013; 81: 1453-9. Crossref
47. Révész D, Rydenhag B, Ben-Menachem E. Complications and safe-
ty of vagus nerve stimulation: 25 years of experience at a single
center. J Neurosurg Pediatr 2016; 18: 97-104. Crossref
48. Morris GL 3rd, Mueller WM, the Vagus Nerve Stimulation Study
Group E01-E05. Long-term treatment with vagus nerve stimulation
in patients with refractory epilepsy. Neurology 1999; 53: 1731-5.
49. Goadsby P, Lipton R, Cady R, Mauskop A, Grosberg A. Non-invasive
vagus nerve stimulation (nVNS) for acute treatment of migraine:
an open-label pilot study (abstract S40.004). Presented at Annual
Meeting of the American Academy of Neurology, 16_23 March
2013, San Diego, CA.
50. Jürgens TP, Leone M. Pearls and pitfalls: neurostimulation in head-
ache. Cephalalgia 2013; 33: 512-25. Crossref
Akdemir et al.
Vagus nerve stimulation Anatol J Cardiol 2016; 16: 804-10
810
From Prof. Dr. Arif Aksit's collection
... Medical device and drug interventions targeting both the extrinsic and intrinsic cardiac nervous system must undergo rigorous validation and safety assessment. ANS-modulating interventions like cardiac sympathetic denervation, renal denervation, vagal stimulation, ganglionated plexi ablation and optogenetics have been employed as investigative tools (Akdemir & Benditt, 2016;Kobayashi et al., 2013;Manolis et al., 2021). Our ivNCC model provides a platform for preclinical testing of novel CCM therapies and other neuromodulation tools. ...
... Our coculture models can be directly used to study multiple cardiac cycles during normal and ischemic events using diseased hiPSC models. Research on the therapeutic modulation of cardiac autonomic tone by electrical stimulation has yielded encouraging early clinical results (Akdemir & Benditt, 2016;De Ferrari et al., 2011;Kobayashi et al., 2013;Schwartz et al., 2008). Direct application of electrical stimuli as in the case of CCM therapy may help stabilize normal neuronal output function, leading to balance between cardiac cholinergic and adrenergic efferent neuronal outputs and thus contributing to suppression of arrhythmias. ...
Article
Full-text available
Two of the most prominent organ systems, the nervous and the cardiovascular systems, are intricately connected to maintain homeostasis in mammals. Recent years have shown tremendous efforts toward therapeutic modulation of cardiac contractility and electrophysiology by electrical stimulation. Neuronal innervation and cardiac ganglia regulation are often overlooked when developing in vitro models for cardiac devices, but it is likely that peripheral nervous system plays a role in the clinical effects. We developed an in vitro neurocardiac coculture (ivNCC) model system to study cardiac and neuronal interplay using human induced pluripotent stem cell (hiPSC) technology. We demonstrated significant expression and colocalization of cardiac markers including troponin, α-actinin, and neuronal marker peripherin in neurocardiac coculture. To assess functional coupling between the cardiomyocytes and neurons, we evaluated nicotine-induced β-adrenergic norepinephrine effect and found beat rate was significantly increased in ivNCC as compared to monoculture alone. The developed platform was used as a nonclinical model for the assessment of cardiac medical devices that deliver nonexcitatory electrical pulses to the heart during the absolute refractory period of the cardiac cycle, that is, cardiac contractility modulation (CCM) therapy. Robust coculture response was observed at 14 V/cm (5 V, 64 mA), monophasic, 2 ms pulse duration for pacing and 20 V/cm (7 V, 90 mA) phase amplitude, biphasic, 5.14 ms pulse duration for CCM. We observed that the CCM effect and kinetics were more pronounced in coculture as compared to cardiac monoculture, supporting a hypothesis that some part of CCM mechanism of action can be attributed to peripheral nervous system stimulation. This study provides novel characterization of CCM effects on hiPSC-derived neurocardiac cocultures. This innervated human heart model can be further extended to investigate arrhythmic mechanisms, neurocardiac safety, and toxicity post-chronic exposure to materials, drugs, and medical devices. We present data on acute CCM electrical stimulation effects on a functional and optimized coculture using commercially available hiPSC-derived cardiomyocytes and neurons. Moreover, this study provides an in vitro human heart model to evaluate neuronal innervation and cardiac ganglia regulation of contractility by applying CCM pulse parameters that closely resemble clinical setting. This ivNCC platform provides a potential tool for investigating aspects of cardiac and neurological device safety and performance.
... First, effects of taVNS on vmHRV may depend on baseline levels of the HRV parameters, since it has been shown that vmHRV is predominantly affected by taVNS in individuals with lower vmHRV to begin with (Clancy et al., 2014). In line with this, our results may not generalize to clinical groups where sympathetic dominance is part of the pathology, such as heart failure or atrial fibrillation (Akdemir & Benditt, 2016;Kaniusas et al., 2019) because of potential differences in baseline indices of cardiac vagal activity (Akdemir & Benditt, 2016). ...
... First, effects of taVNS on vmHRV may depend on baseline levels of the HRV parameters, since it has been shown that vmHRV is predominantly affected by taVNS in individuals with lower vmHRV to begin with (Clancy et al., 2014). In line with this, our results may not generalize to clinical groups where sympathetic dominance is part of the pathology, such as heart failure or atrial fibrillation (Akdemir & Benditt, 2016;Kaniusas et al., 2019) because of potential differences in baseline indices of cardiac vagal activity (Akdemir & Benditt, 2016). ...
Article
Full-text available
Non-invasive brain stimulation techniques, such as transcutaneous auricular vagus nerve stimulation (taVNS), have considerable potential for clinical use. Beneficial effects of taVNS have been demonstrated on symptoms in patients with mental or neurological disorders as well as transdiagnostic dimensions, including mood and motivation. However, since taVNS research is still an emerging field, the underlying neurophysiological processes are not yet fully understood, and the replicability of findings on biomarkers of taVNS effects has been questioned. The objective of this analysis was to synthesize the current evidence concerning the effects of taVNS on vagally mediated heart rate variability (vmHRV), a candidate biomarker that has, so far, received most attention in the field. We performed a living Bayesian random effects meta-analysis. To keep the synthesis of evidence transparent and up to date as new studies are being published, we developed a Shiny web app that regularly incorporates new results and enables users to modify study selection criteria to evaluate the robustness of the inference across potential confounds. Our analysis focuses on 16 single-blind studies comparing taVNS versus sham in healthy participants. The meta-analysis provides strong evidence for the null hypothesis (g = 0.014, CIshortest = [-0.103, 0.132], BF01 = 24.678), indicating that acute taVNS does not alter vmHRV compared to sham. To conclude, there is no support for the hypothesis that vmHRV is a robust biomarker for acute taVNS. By increasing transparency and timeliness, the concept of living meta-analyses can lead to transformational benefits in emerging fields such as non-invasive brain stimulation.
... First, taVNS serves as a potential treatment for patient groups where sympathetic dominance is part of the pathology, such as heart failure or atrial fibrillation (Akdemir & Benditt, 2016;Kaniusas et al., 2019). However, assuming generalizability of the results to clinical populations is problematic because of potential differences in baseline indices of cardiac vagal activity (Akdemir & Benditt, 2016). ...
... First, taVNS serves as a potential treatment for patient groups where sympathetic dominance is part of the pathology, such as heart failure or atrial fibrillation (Akdemir & Benditt, 2016;Kaniusas et al., 2019). However, assuming generalizability of the results to clinical populations is problematic because of potential differences in baseline indices of cardiac vagal activity (Akdemir & Benditt, 2016). Second, except for a few studies (De Couck et al., 2017;Sclocco et al., 2020), earlobe sham is chosen as control condition, because it evokes a similar sensation to the participant, without actively targeting the vagus nerve (Frangos et al., 2015). ...
Preprint
Full-text available
Non-invasive brain stimulation techniques, such as transcutaneous auricular vagus nerve stimulation (taVNS), have considerable potential for clinical use. Beneficial effects of taVNS have been demonstrated on symptoms in patients with mental or neurological disorders as well as transdiagnostic dimensions, including mood and motivation. However, since taVNS research is still an emerging field, the underlying neurophysiological processes are not yet fully understood, and the replicability of findings on biomarkers of taVNS effects has been questioned. Here, we perform a living Bayesian random effects meta-analysis to synthesize the current evidence concerning the effects of taVNS on heart rate variability (HRV), a candidate biomarker that has, so far, received most attention in the field. To keep the synthesis of evidence transparent and up to date as new studies are being published, we developed a Shiny web app that regularly incorporates new results and enables users to modify study selection criteria to evaluate the robustness of the inference across potential confounds. Our analysis focuses on 17 single-blind studies comparing taVNS versus sham in healthy participants. These newly synthesized results provide strong evidence for the null hypothesis ( g = 0.011, CI shortest = [-0.103, 0.125], BF 01 = 25.587), indicating that acute taVNS does not alter HRV compared to sham. To conclude, based on a synthesis of the available evidence to date, there is no support for the hypothesis that HRV is a robust biomarker for acute taVNS. By increasing transparency and timeliness, we believe that the concept of living meta-analyses can lead to transformational benefits in emerging fields such as non-invasive brain stimulation.
... Tragus nerve stimulation. One of the main drawbacks of vagus stimulation is the invasive nature of this therapy, which is accompanied by surgical risks and low patient tolerance [110]. Low-level tragus stimulation (LLTS) is a non-invasive transcutaneous approach that may influence autonomic function by stimulating the auricular branch of the vagus nerve (ABVN) [111]. ...
Article
Full-text available
Heart failure (HF) is a progressively deteriorating medical condition that significantly reduces both the patients’ life expectancy and quality of life. Even though real progress was made in the past decades in the discovery of novel pharmacological treatments for HF, the prevention of premature deaths has only been marginally alleviated. Despite the availability of a plethora of pharmaceutical approaches, proper management of HF is still challenging. Thus, a myriad of experimental and clinical studies focusing on the discovery of new and provocative underlying mechanisms of HF physiopathology pave the way for the development of novel HF therapeutic approaches. Furthermore, recent technological advances made possible the development of various interventional techniques and device-based approaches for the treatment of HF. Since many of these modern approaches interfere with various well-known pathological mechanisms in HF, they have a real ability to complement and or increase the efficiency of existing medications and thus improve the prognosis and survival rate of HF patients. Their promising and encouraging results reported to date compel the extension of heart failure treatment beyond the classical view. The aim of this review was to summarize modern approaches, new perspectives, and future directions for the treatment of HF.
... A recently published meta-analysis of RCTs with invasive VNS (iVNS) found that the intervention leads to a significant improvement in NYHA functional classification, quality of life, 6-min walking test distance, and NT-proBNP levels; however, they found that there was no difference in mortality (Sant'Anna et al., 2021). While iVNS has shown interesting results as an alternative therapeutic approach for heart failure, the invasiveness and cost of the surgical procedure, and poor patient tolerance limit the clinical applicability (Akdemir and Benditt, 2016). ...
Article
Full-text available
The heart is electrically and mechanically controlled by the autonomic nervous system, which consists of both the sympathetic and parasympathetic systems. It has been considered that the sympathetic and parasympathetic nerves regulate the cardiomyocytes’ performance independently; however, recent molecular biology approaches have provided a new concept to our understanding of the mechanisms controlling the diseased heart through the plasticity of the autonomic nervous system. Studies have found that cardiac sympathetic nerve fibers in hypertrophic ventricles strongly express an immature neuron marker and simultaneously cause deterioration of neuronal cellular function. This phenomenon was explained by the rejuvenation of cardiac sympathetic nerves. Moreover, heart failure and myocardial infarction have been shown to cause cholinergic trans-differentiation of cardiac sympathetic nerve fibers via gp130-signaling cytokines secreted from the failing myocardium, affecting cardiac performance and prognosis. This phenomenon is thought to be one of the adaptations that prevent the progression of heart disease. Recently, the concept of using device-based neuromodulation therapies to attenuate sympathetic activity and increase parasympathetic (vagal) activity to treat cardiovascular disease, including heart failure, was developed. Although several promising preclinical and pilot clinical studies using these strategies have been conducted, the results of clinical efficacy vary. In this review, we summarize the current literature on the plasticity of cardiac sympathetic nerves and propose potential new therapeutic targets for heart disease.
... VNS application extends beyond neurological diseases: a growing body of clinical evidence from human patients indicates its suitability for treatment of chronic heart failure (90,91) or inflammatory diseases such as Crohn's disease (92,93) or rheumatoid arthritis (94,95). Recently, nVNS has been proposed and applied to patients with respiratory symptoms of COVID-19 to modulate their inflammatory response (96)(97)(98). ...
Article
Full-text available
Modulation of neuronal activity for seizure control using various methods of neurostimulation is a rapidly developing field in epileptology, especially in treatment of refractory epilepsy. Promising results in human clinical practice, such as diminished seizure burden, reduced incidence of sudden unexplained death in epilepsy, and improved quality of life has brought neurostimulation into the focus of veterinary medicine as a therapeutic option. This article provides a comprehensive review of available neurostimulation methods for seizure management in drug-resistant epilepsy in canine patients. Recent progress in non-invasive modalities, such as repetitive transcranial magnetic stimulation and transcutaneous vagus nerve stimulation is highlighted. We further discuss potential future advances and their plausible application as means for preventing epileptogenesis in dogs.
... In particular, several pathophysiological aspects of stroke (e.g., neuroinflammation) are influenced by the activity of the vagus nerve [125]. In addition to depression and epilepsy, vagal stimulators have been already used in other chronic conditions such as chronic pain, refractory migraine and cardiovascular diseases, testifying to their safety profile [126,127]. Moreover, recent preclinical research on the cholinergic anti-inflammatory pathway is expanding the use of vagus nerve stimulation (VNS) to target neuroinflammation in neurological disorders such as Alzheimer's disease, Parkinson's disease and traumatic brain injuries [128][129][130]. ...
Article
Full-text available
Ischemic stroke is a worldwide major cause of mortality and disability and has high costs in terms of health-related quality of life and expectancy as well as of social healthcare resources. In recent years, starting from the bidirectional relationship between autonomic nervous system (ANS) dysfunction and acute ischemic stroke (AIS), researchers have identified prognostic factors for risk stratification, prognosis of mid-term outcomes and response to recanalization therapy. In particular, the evaluation of the ANS function through the analysis of heart rate variability (HRV) appears to be a promising non-invasive and reliable tool for the management of patients with AIS. Furthermore, preclinical molecular studies on the pathophysiological mechanisms underlying the onset and progression of stroke damage have shown an extensive overlap with the activity of the vagus nerve. Evidence from the application of vagus nerve stimulation (VNS) on animal models of AIS and on patients with chronic ischemic stroke has highlighted the surprising therapeutic possibilities of neuromodulation. Preclinical molecular studies highlighted that the neuroprotective action of VNS results from anti-inflammatory, antioxidant and antiapoptotic mechanisms mediated by α7 nicotinic acetylcholine receptor. Given the proven safety of non-invasive VNS in the subacute phase, the ease of its use and its possible beneficial effect in hemorrhagic stroke as well, human studies with transcutaneous VNS should be less challenging than protocols that involve invasive VNS and could be the proof of concept that neuromodulation represents the very first therapeutic approach in the ultra-early management of stroke.
... It is used by sending mild electrical stimulation through the skin to activate the vagus nerve from outside the body. It is programmed for stimulation in cycles for 2 min, and one treatment consists of three cycles (Akdemir and Benditt, 2016;Mwamburi et al., 2017). ...
Article
Full-text available
Autonomic imbalance plays a crucial role in the genesis and maintenance of cardiac disorders. Approaches to maintain sympatho-vagal balance in heart diseases have gained great interest in recent years. Emerging therapies However, certain types of emerging therapies including direct electrical stimulation and nerve denervation require invasive implantation of a generator and a bipolar electrode subcutaneously or result in autonomic nervous system (ANS) damage, inevitably increasing the risk of complications. More recently, non-invasive neuromodulation approaches have received great interest in ANS modulation. Non-invasive approaches have opened new fields in the treatment of cardiovascular diseases. Herein, we will review the protective roles of non-invasive neuromodulation techniques in heart diseases, including transcutaneous auricular vagus nerve stimulation, electromagnetic field stimulation, ultrasound stimulation, autonomic modulation in optogenetics, and light-emitting diode and transcutaneous cervical vagus nerve stimulation (gammaCore).
Article
Objectives Clinical outcomes following myocardial ischemia–reperfusion (I/R) injury are strongly related to the intensity and duration of inflammation. The splenic nerve (SpN) is indispensable for the anti-inflammatory reflex. This study aimed to investigate whether splenic nerve stimulation (SpNS) plays a cardioprotective role in myocardial I/R injury and the potential underlying mechanism. Methods Sprague-Dawley rats were randomly divided into four groups: sham group, I/R group, SpNS group, and I/R plus SpNS group. The highest SpNS intensity that did not influence heart rate was identified, and SpNS at this intensity was used as the subthreshold stimulus. Continuous subthreshold SpNS was applied for 1 h before ligation of the left coronary artery for 45 min. After 72 h of reperfusion, samples were collected for analysis. Results SpN activity and splenic concentrations of cholinergic anti-inflammatory pathway (CAP)-related neurotransmitters were significantly increased by SpNS. The infarct size, oxidative stress, sympathetic tone, and the levels of proinflammatory cytokines, including TNF-α, IL-1β, and IL-6, were significantly reduced in rats subjected to subthreshold SpNS after myocardial I/R injury compared with those subjected to I/R injury alone. Conclusions Subthreshold SpNS ameliorates myocardial damage, the inflammatory response, and cardiac remodelling induced by myocardial I/R injury via neuroimmunomodulation of proinflammatory factor levels. SpNS is a potential therapeutic strategy for the treatment of myocardial I/R injury.
Article
Full-text available
Objectives After 20 years of development, there is a confusion in the nomenclature of transcutaneous stimulation of the auricular branch of the vagus nerve (ABVN). We performed a systematic review of transcutaneous stimulation of ABVN in nomenclature. Materials and Methods A systematic search of the literature was carried out, using the bibliographic search engines PubMed. The search covered articles published up until June 11, 2020. We recorded the full nomenclature and abbreviated nomenclature same or similar to transcutaneous stimulation of ABVN in the selected eligible studies, as well as the time and author information of this nomenclature. Results From 261 studies, 67 full nomenclatures and 27 abbreviated nomenclatures were finally screened out, transcutaneous vagus nerve stimulation and tVNS are the most common nomenclature, accounting for 38.38% and 42.06%, respectively. In a total of 97 combinations of full nomenclatures and abbreviations, the most commonly used nomenclature for the combination of transcutaneous vagus nerve stimulation and tVNS, accounting for 30.28%. Interestingly, the combination of full nomenclatures and abbreviations is not always a one‐to‐one relationship, there are ten abbreviated nomenclatures corresponding to transcutaneous vagus nerve stimulation, and five full nomenclatures corresponding to tVNS. In addition, based on the analysis of the usage habits of nomenclature in 21 teams, it is found that only three teams have fixed habits, while other different teams or the same team do not always use the same nomenclature in their paper. Conclusions The phenomenon of confusion in the nomenclature of transcutaneous stimulation of ABVN is obvious and shows a trend of diversity. The nomenclature of transcutaneous stimulation of ABVN needs to become more standardized in the future.
Article
Full-text available
Vagus nerve stimulation (VNS) is effective in refractory epilepsy and depression and is being investigated in heart failure, headache, gastric motility disorders and asthma. The first VNS device required surgical implantation of electrodes and a stimulator. Adverse events (AEs) are generally associated with implantation or continuous on-off stimulation. Infection is the most serious implantation-associated AE. Bradycardia and asystole have also been described during implantation, as has vocal cord paresis, which can last up to 6 months and depends on surgical skill and experience. The most frequent stimulation-associated AEs include voice alteration, paresthesia, cough, headache, dyspnea, pharyngitis and pain, which may require a decrease in stimulation strength or intermittent or permanent device deactivation. Newer non-invasive VNS delivery systems do not require surgery and permit patient-administered stimulation on demand. These non-invasive VNS systems improve the safety and tolerability of VNS, making it more accessible and facilitating further investigations across a wider range of uses. © 2015 The Authors. European Journal of Neurology published by John Wiley & Sons Ltd on behalf of European Academy of Neurology.
Article
Full-text available
Aim The neural cardiac therapy for heart failure (NECTAR-HF) was a randomized sham-controlled trial designed to evaluate whether a single dose of vagal nerve stimulation (VNS) would attenuate cardiac remodelling, improve cardiac function and increase exercise capacity in symptomatic heart failure patients with severe left ventricular (LV) systolic dysfunction despite guideline recommended medical therapy. Methods Patients were randomized in a 2 : 1 ratio to receive therapy (VNS ON) or control (VNS OFF) for a 6-month period. The primary endpoint was the change in LV end systolic diameter (LVESD) at 6 months for control vs. therapy, with secondary endpoints of other echocardiography measurements, exercise capacity, quality-of-life assessments, 24-h Holter, and circulating biomarkers. Results Of the 96 implanted patients, 87 had paired datasets for the primary endpoint. Change in LVESD from baseline to 6 months was −0.04 ± 0.25 cm in the therapy group compared with −0.08 ± 0.32 cm in the control group (P = 0.60). Additional echocardiographic parameters of LV end diastolic dimension, LV end systolic volume, left ventricular end diastolic volume, LV ejection fraction, peak V02, and N-terminal pro-hormone brain natriuretic peptide failed to show superiority compared to the control group. However, there were statistically significant improvements in quality of life for the Minnesota Living with Heart Failure Questionnaire (P = 0.049), New York Heart Association class (P = 0.032), and the SF-36 Physical Component (P = 0.016) in the therapy group. Conclusion Vagal nerve stimulation as delivered in the NECTAR-HF trial failed to demonstrate a significant effect on primary and secondary endpoint measures of cardiac remodelling and functional capacity in symptomatic heart failure patients, but quality-of-life measures showed significant improvement.
Article
OBJECTIVE The goal of this paper was to investigate surgical and hardware complications in a longitudinal retrospective study. METHODS The authors of this registry study analyzed the surgical and hardware complications in 247 patients who underwent the implantation of a vagus nerve stimulation (VNS) device between 1990 and 2014. The mean follow-up time was 12 years. RESULTS In total, 497 procedures were performed for 247 primary VNS implantations. Complications related to surgery occurred in 8.6% of all implantation procedures that were performed. The respective rate for hardware complications was 3.7%. Surgical complications included postoperative hematoma in 1.9%, infection in 2.6%, vocal cord palsy in 1.4%, lower facial weakness in 0.2%, pain and sensory-related complications in 1.4%, aseptic reaction in 0.2%, cable discomfort in 0.2%, surgical cable break in 0.2%, oversized stimulator pocket in 0.2%, and battery displacement in 0.2% of patients. Hardware-related complications included lead fracture/malfunction in 3.0%, spontaneous VNS turn-on in 0.2%, and lead disconnection in 0.2% of patients. CONCLUSIONS VNS implantation is a relatively safe procedure, but it still involves certain risks. The most common complications are postoperative hematoma, infection, and vocal cord palsy. Although their occurrence rates are rather low at about 2%, these complications may cause major suffering and even be life threatening. To reduce complications, it is important to have a long-term perspective. The 25 years of follow-up of this study is of great strength considering that VNS can be a life-long treatment for many patients. Thus, it is important to include repeated surgeries such as battery and lead replacements, given that complications also may occur with these surgeries.
Article
Background: beta-Blockade-induced benefit in heart failure (HF) could be related to baseline heart rate and treatment-induced heart rate reduction, but no such relationships have been demonstrated. Methods and results: In CIBIS II, we studied the relationships between baseline heart rate (BHR), heart rate changes at 2 months (HRC), nature of cardiac rhythm (sinus rhythm or atrial fibrillation), and outcomes (mortality and hospitalization for HF). Multivariate analysis of CIBIS II showed that in addition to beta-blocker treatment, BHR and HRC were both significantly related to survival and hospitalization for worsening HF, the lowest BHR and the greatest HRC being associated with best survival and reduction of hospital admissions. No interaction between the 3 variables was observed, meaning that on one hand, HRC-related improvement in survival was similar at all levels of BHR, and on the other hand, bisoprolol-induced benefit over placebo for survival was observed to a similar extent at any level of both BHR and HRC. Bisoprolol reduced mortality in patients with sinus rhythm (relative risk 0.58, P:<0.001) but not in patients with atrial fibrillation (relative risk 1.16, P:=NS). A similar result was observed for cardiovascular mortality and hospitalization for HF worsening. Conclusions: BHR and HRC are significantly related to prognosis in heart failure. beta-Blockade with bisoprolol further improves survival at any level of BHR and HRC and to a similar extent. The benefit of bisoprolol is questionable, however, in patients with atrial fibrillation.
Article
VNS, as delivered by the NCP system, is an FDA-approved, well-tolerated, effective intervention for a substantial proportion of patients with treatment-resistant epilepsy. Its longer term safety and tolerability are well documented in these patients. There are several clinical and theoretical reasons to think that VNS might be an effective antidepressant or mood stabilizer. A recent open, uncontrolled trial provided encouraging results. Most patients who responded acutely have maintained the benefits. Some subjects responded after exiting the acute study, while continuing to receive VNS in an uncontrolled follow-up. These results are similar to the increasing benefits with VNS over the longer term observed in patients with epilepsy. Major questions ranging from the need to establish efficacy in a controlled, comparative trial; determining indicators of who will respond; documentation of long-term efficacy; studies of cost recovery; and where VNS fits into the overall disease management scheme for depression deserve careful study.
Article
Ictal bradycardia and ictal asystole, although well known to neurologists, have received relatively little attention in the cardiology community. Consequently, in certain instances, the true pathogenesis for heart rate slowing (ie, epilepsy) may be missed, and the bradyarrhythmia incorrectly attributed to other causes. Technically, both ictal bradycardia and asystole are more precisely termed ictal-induced cardiac bradyarrhythmia or asystole. Most often the neurological trigger is a complex partial seizure, which may or may not become generalized. 1 Further, if sufficiently severe, the ictal-induced bradyarrhythmia temporarily impairs both cerebral perfusion and cortical function; the result has the dual effect of terminating the seizure, while at the same time triggering syncope with consequent loss of consciousness and postural tone. In essence, a complex partial seizure patient may manifest both seizure and syncope features during the same episode. Article see p 159