Fact-checked by Grok 2 weeks ago

Vagus nerve stimulation

Vagus nerve stimulation (VNS) is a therapy that delivers controlled electrical impulses to the , the longest cranial nerve (cranial nerve X) in the and the principal component of the , which regulates vital functions including , , and respiratory activity. This treatment modulates brain activity by stimulating afferent fibers of the nerve, primarily targeting the left cervical vagus trunk via an implantable or non-invasive external devices, and is FDA-approved for , , stroke rehabilitation, cluster headaches, and . The origins of VNS trace back to the late , but modern invasive VNS was pioneered in the 1980s through research demonstrating its effects, leading to FDA approval in 1997 for adults with drug-resistant and expansion to children aged 4 and older in 2017. In 2005, it received approval for after at least four failed trials, with response rates of 27–46% in long-term studies. The therapy's mechanism involves projecting signals to the nucleus tractus solitarius in the , which influences neurotransmitter systems (e.g., increasing serotonin and norepinephrine), promotes neural plasticity via (BDNF), and activates the anti-inflammatory pathway to reduce . Surgical implantation, performed under general as an outpatient , places the device subcutaneously in the chest with leads coiled around the left to minimize cardiac effects; stimulation parameters (e.g., 30 seconds on, 5 minutes off) are programmable, and patients can activate extra bursts using a handheld . Beyond core indications, VNS has expanded applications, including FDA approval in 2015 for treatment via a surgically implanted device that promotes and in 2021 for upper extremity motor deficits post-stroke when paired with . Non-invasive variants, such as transcutaneous auricular VNS (taVNS) and transcutaneous VNS (tcVNS), apply stimulation to the or without and show efficacy in reducing severity, frequency (up to 41% in some trials), and depressive symptoms, with ongoing research into and . Common side effects include hoarseness, cough, and throat discomfort during stimulation, which often diminish over time, though rare risks encompass , vocal cord , and device malfunction requiring battery replacement every 3–5 years. Emerging closed-loop systems and optimized protocols promise greater personalization and accessibility, positioning VNS as a versatile tool in and psychiatry for patients with limited treatment options.

Fundamentals

The vagus nerve

The , also known as cranial nerve X, is the tenth paired cranial nerve and the longest in the , extending from the to the . It originates in the , specifically from the dorsal motor nucleus for parasympathetic efferents and the for branchial motor fibers, with sensory components arising from the inferior ganglion. As a bilateral structure, the right and left vagus nerves emerge from the skull via the and descend through the neck within the , alongside the and . In the neck, the vagus nerve gives rise to several branches, including the auricular branch from the superior ganglion, which provides sensory innervation to the external auditory canal and posterior auricle; the pharyngeal branch from the inferior ganglion, contributing to the pharyngeal plexus for motor control of pharyngeal and soft palate muscles (except the tensor veli palatini); and the superior laryngeal nerve, which divides into internal and external branches to supply sensory innervation to the larynx above the vocal cords and motor innervation to the cricothyroid muscle, respectively. The recurrent laryngeal nerve, a major branch, loops under the right subclavian artery on the right side and the aortic arch on the left, ascending to innervate all intrinsic laryngeal muscles except the cricothyroid, facilitating phonation and airway protection. As the nerves enter the thorax, they pass posterior to the root of the lung, forming the pulmonary plexus with sympathetic fibers to regulate bronchial tone and secretion; cardiac branches arise to form the cardiac plexus, providing parasympathetic innervation to slow heart rate via the sinoatrial and atrioventricular nodes. Continuing into the abdomen through the esophageal hiatus, the vagus nerves form the esophageal plexus before separating into anterior and posterior trunks, which branch into gastric, celiac, and hepatic divisions to innervate the stomach, intestines, liver, pancreas, and other viscera, controlling gastrointestinal motility and secretion. Physiologically, the serves as a primary component of the parasympathetic division of the , exerting inhibitory control over visceral functions to promote "rest and digest" states. It regulates by releasing onto cardiac ganglia, reducing sinoatrial node firing; modulates through pulmonary branches that influence bronchial diameter and mucus production; and governs digestion by stimulating secretion, , and pancreatic enzyme release via connections. Sensory functions are extensive, with visceral afferents conveying information from in the and , chemoreceptors detecting blood gas levels, and mechanoreceptors in the signaling satiety and discomfort to the . The nerve's fiber composition is predominantly afferent, with approximately 80% sensory fibers transmitting interoceptive signals from the viscera to the and 20% efferent fibers mediating parasympathetic outflow. Notably, the participates in the cholinergic pathway, where efferent signals release to inhibit production in macrophages via alpha-7 nicotinic receptors, thereby attenuating . Vagal tone, a measure of parasympathetic nervous system activity, is commonly assessed through (HRV), particularly the high-frequency component reflecting respiratory , which correlates with vagal modulation of cardiac function. Evolutionarily, the vagus nerve represents a conserved element of the , with its myelinated ventral branch emerging in mammals to support behaviors through integration with and vocal motor pathways, enhancing survival via coordinated physiological responses to environmental cues.

Stimulation principles

Vagus nerve stimulation (VNS) operates on the biophysical principle of delivering controlled electrical pulses to the afferent fibers of the , primarily the myelinated A and B fibers, to evoke action potentials that propagate centrally without causing direct nerve damage. These pulses are characterized by specific parameters optimized for therapeutic and : typical pulse widths range from 250 to 500 μs, frequencies from 20 to 30 Hz, output currents from 0.25 to 3.5 mA, and a of 30 seconds on followed by 5 minutes off to prevent overstimulation and accommodate battery life in implanted systems. The electrical current recruits fibers in a size-dependent manner, with larger A and B fibers activating at lower intensities to modulate autonomic and sensory pathways, while smaller unmyelinated C fibers require higher currents and are generally avoided in standard protocols to minimize pain or off-target effects. Targeting specificity is achieved by selecting the stimulation site and side of the , with the left branch preferred for most applications due to its predominantly afferent composition and reduced risk of cardiac side effects like , which are more prominent with right-sided owing to greater efferent innervation of the . Selective activation focuses on afferent A and B fibers to promote central signaling while sparing efferent and C fibers, often through design that encircles the or applies surface to accessible branches. VNS encompasses invasive and non-invasive types, differing in delivery method and accessibility. Invasive VNS involves direct contact via a surgically implanted cuff around the , enabling precise, chronic stimulation with programmable . Non-invasive approaches, such as transcutaneous VNS (tVNS), apply electrical pulses through the skin to peripheral branches like the auricular vagus in the external ear, offering a less intrusive with similar ranges but lower . Stimulation paradigms include open-loop systems, which deliver pulses on a fixed schedule regardless of physiological state, and closed-loop systems, which trigger pulses in response to detected biomarkers like onset for more targeted intervention. The basic neurophysiological response to VNS involves the generation and orthodromic propagation of action potentials along afferent fibers to the , where they primarily in the tractus solitarius (NTS), initiating a cascade of neural modulation across connected nuclei. This activation of NTS neurons, particularly those receiving myelinated vagal input, occurs at therapeutic intensities and sustains central effects without requiring supramedullary pathways for initial relay.

Therapeutic Applications

Epilepsy

Vagus nerve stimulation (VNS) received initial FDA approval in 1997 as an adjunctive therapy for reducing the frequency of partial-onset seizures in patients with refractory to antiepileptic medications. This approval was based on clinical evidence demonstrating its efficacy in adults and adolescents aged 12 years and older. Subsequent expansions in 2017 extended the indication to children as young as 4 years old with focal seizures inadequately controlled by medications, positioning VNS as a long-term, non-pharmacological option alongside ongoing antiepileptic drug therapy. Patient selection for VNS in emphasizes individuals with drug-resistant focal s, typically defined as failure to achieve adequate control after trials of at least two appropriately chosen and dosed antiepileptic drugs, either as monotherapies or in combination. Common criteria include a frequency exceeding three per month, primarily partial-onset s, and confirmation that s are not psychogenic through video-EEG when necessary. Contraindications include a history of bilateral or left cervical , as this precludes safe implantation and stimulation, and cardiac conduction abnormalities that could be exacerbated by vagal effects. For instance, patients with bilateral vocal cord are generally excluded due to heightened risk of respiratory complications from stimulation-induced laryngeal effects. Treatment protocols for VNS implantation involve surgical placement of a in the chest, connected to an wrapped around the left , followed by a gradual phase to optimize efficacy while minimizing s. Stimulation begins at low settings, such as 0.25 mA output current, 250 μs , 20-30 Hz , and a 7-second on/18- to 60-second off , with incremental increases of 0.25 mA every 1-2 weeks over an initial 2-4 week period until a tolerable therapeutic dose (typically 1-2.5 mA) is reached. Patients or caregivers can use a handheld to deliver on-demand stimulation bursts for acute interruption or as a boost during aura perception, providing an additional layer of control integrated with daily antiepileptic regimens. Long-term management includes periodic outpatient adjustments based on logs and tolerance, often in conjunction with specialists to complement or other interventions like ketogenic diets. Clinical response to VNS is commonly assessed by the proportion of patients achieving at least a 50% reduction in seizure frequency, with studies showing this outcome in 50-60% of individuals after two years of therapy, alongside improvements in seizure severity and . This adjunctive role allows VNS to enhance overall seizure management without replacing medications, particularly in pediatric cases where developmental benefits may accrue over time.

Treatment-resistant depression

Vagus nerve stimulation (VNS) received U.S. (FDA) approval in 2005 as an adjunctive long-term treatment for chronic or recurrent in adults aged 18 years or older who are experiencing a major depressive episode and have not responded adequately to at least four treatments. This indication encompasses both unipolar and , positioning VNS as a option for cases where standard has proven insufficient. Clinical trials establishing this approval utilized reduction in Hamilton Depression Rating Scale (HAMD-17) scores as the primary outcome measure to assess symptom improvement. Patient selection for VNS emphasizes individuals with chronic depression persisting for more than two years or recurrent episodes, characterized by severe symptoms such as elevated baseline HAMD scores typically exceeding 20. Candidates must demonstrate treatment resistance through documented failure of multiple interventions, often quantified by an Antidepressant Treatment History Form (ATHF) score of at least 3. Exclusion criteria include acute suicidality, defined as recent suicide attempts or high imminent risk within the past 12 months, to mitigate perioperative and post-implant risks. Psychiatric evaluation ensures suitability, prioritizing those with stable but refractory illness over acutely unstable presentations. Post-implantation, VNS involves continuous pulsed electrical stimulation of the left via an implanted , with initial parameters set at 0.25–1.0 mA output current, 250–500 μs , and a 30-second on/5-minute off cycle, adjustable based on tolerability and response. Therapeutic benefits often exhibit a delayed onset, with meaningful symptom reduction requiring up to 12 months of ongoing stimulation, distinguishing VNS from faster-acting interventions. Treatment protocols integrate VNS with continued and to optimize outcomes, as the device functions adjunctively without replacing conventional care. A key advantage of VNS lies in its potential for sustained remission, with long-term observational data indicating 20–30% of patients achieving durable response rates beyond one year, including reduced relapse and improved . These outcomes reflect cumulative benefits over time, with remission defined as HAMD-17 scores below 7 or 10. The anti-inflammatory pathways activated by VNS, which suppress proinflammatory cytokines, may underlie these mood-stabilizing effects in treatment-resistant cases.

Migraine and cluster headaches

Non-invasive vagus nerve stimulation (nVNS), particularly via devices like gammaCore, has been approved for treating primary headache disorders such as and . The U.S. (FDA) first cleared gammaCore in April 2017 for the acute of associated with episodic in adults. This clearance was expanded in January 2018 to include the acute of associated with headache in adult patients, based on results from the randomized PRESTO trial demonstrating superior efficacy over sham stimulation. Further expansions occurred in November 2018 for adjunctive preventive of in adults and in subsequent years for preventive use in , including chronic forms, for patients aged 12 and older. These approvals target trigeminal-autonomic cephalalgias and phenotypes, offering a non-pharmacological option that modulates autonomic pathways to reduce vascular and inflammatory components of cephalic . Patient selection for nVNS in migraine and cluster headache emphasizes distinguishing episodic from chronic forms to optimize outcomes. For cluster headache, nVNS shows greater efficacy in episodic cases (with attack periods lasting 7-365 days per year) compared to chronic forms (attacks persisting over a year without remission), as evidenced by higher response rates in pivotal trials like ACT1 for episodic patients. In migraine, both episodic (fewer than 15 headache days per month) and chronic (15 or more headache days per month) subtypes are eligible, though preventive strategies are particularly suited to chronic migraine to reduce attack frequency. Contraindications include active implantable devices such as pacemakers or defibrillators, due to potential electromagnetic interference; severe carotid artery disease or atherosclerosis; recent neck surgery or injury; and use in patients under 12 years old or during pregnancy, as safety data are limited in these groups. Candidates are typically adults with inadequately controlled symptoms despite standard therapies, excluding those with secondary headaches or unstable cardiovascular conditions. Treatment protocols for nVNS in these headaches involve self-administered transcutaneous stimulation applied to the cervical branch of the vagus nerve on the neck using a handheld device. For acute migraine or cluster attacks, stimulation is initiated at pain onset or during the aura phase, consisting of two consecutive 2-minute doses (120 seconds each) on the same or alternating sides of the neck, with conductive gel to ensure contact; if pain persists, additional treatments can be applied up to three times per episode, spaced 10-15 minutes apart, not exceeding 30 stimulations daily. Preventive protocols for chronic migraine or cluster headache recommend twice-daily sessions—once within an hour of waking and once at bedtime—each comprising two 2-minute stimulations on the affected side, aiming to reduce overall attack frequency over weeks to months. These regimens are portable and drug-free, allowing integration with rescue medications like triptans without known interactions. Clinical response metrics highlight nVNS efficacy, particularly for acute treatment, with pain freedom at 2 hours post-stimulation reported in approximately 30% of attacks in the PRESTO (versus 20% with ), establishing meaningful relief in a subset of patients comparable to some oral therapies. For episodic , acute nVNS achieves pain freedom in 15-20% of attacks within 15 minutes, rising to over 30% by 2 hours in responder analyses from the ACT1 and ACT2 studies, though rates are lower (around 10-15%) in chronic . Preventive use in chronic reduces monthly days by 20-30% in open-label extensions, with sustained benefits observed in up to 40% of patients after 3 months. These outcomes underscore nVNS as a targeted for headache phenotypes responsive to autonomic .

Chronic pain

Vagus nerve stimulation (VNS) is considered an investigational and off-label treatment for syndromes such as and (CRPS), where it is not recommended as a first-line due to limited regulatory approvals and ongoing needs. In , a centralized disorder characterized by widespread musculoskeletal pain and , VNS aims to address underlying autonomic dysregulation and central sensitization, with clinical trials exploring both invasive and transcutaneous approaches. For CRPS, a condition involving disproportionate regional pain following injury, VNS is hypothesized to mitigate via the anti-inflammatory pathway, though evidence remains preliminary and it is rarely used clinically. These applications position VNS as an adjunctive option after failure of conventional treatments like . Patient selection for VNS in emphasizes individuals with centralized or components exhibiting involvement, such as altered or sympathetic overactivity common in and CRPS. Ideal candidates are adults over 18 years with a confirmed per established criteria (e.g., American College of Rheumatology guidelines for ) who have not responded to first-line interventions, including opioids, antidepressants, or anticonvulsants. This refractory status ensures VNS is targeted at those with persistent symptoms despite multidisciplinary management, minimizing risks in lower-severity cases. Treatment protocols for VNS in typically involve low-intensity electrical stimulation to minimize side effects while promoting effects, often delivered via non-invasive transcutaneous auricular devices targeting the auricular branch of the . Sessions may last 20-60 minutes, 1-2 times daily, with parameters such as low-frequency pulses (e.g., 20-25 Hz) and currents below 1 , adjusted based on patient tolerance. Integration with or exercise enhances outcomes, as combined approaches have shown improved pain modulation in trials. Clinical outcomes demonstrate moderate reductions, with visual analog (VAS) scores decreasing by 20-50% in responsive patients, attributed to descending inhibitory pathways that dampen nociceptive signaling in the . In fibromyalgia cohorts, meta-analyses report effect sizes around 0.42 for , with some studies noting VAS improvements of approximately 2.5-3 points on a 10-point after regular sessions. For in CRPS, similar reductions occur through mechanisms, though larger trials are needed to confirm durability. This effect may involve brief modulation of neurotransmitters like serotonin in the .

Cardiovascular conditions

Vagus nerve stimulation (VNS) has been investigated as a therapeutic option for cardiovascular conditions, particularly with reduced (HFrEF) and (AF), by modulating autonomic balance to enhance parasympathetic tone and reduce sympathetic overdrive. In HFrEF, VNS targets patients with New York Heart Association (NYHA) class III or IV symptoms despite optimal medical therapy, typically those with left ventricular (LVEF) ≤40% and no recent or cardiac surgery. Patient selection excludes individuals with bradyarrhythmias, chronic AF, or severe comorbidities such as renal or hepatic failure to minimize procedural risks. For AF, candidates include those with paroxysmal episodes or postoperative risk, selected based on autonomic imbalance contributing to arrhythmogenesis. Treatment protocols for HFrEF generally involve left cervical VNS implantation, delivering intermittent electrical pulses (e.g., 3.5-5.5 mA, 10-20 Hz) to augment parasympathetic activity and improve cardiac remodeling. In contrast, right-sided low-frequency stimulation (e.g., <5 Hz) is explored for AF to restore autonomic equilibrium and suppress arrhythmic substrates without excessive bradycardia. These approaches leverage the vagus nerve's role in heart rate control, as detailed in foundational descriptions of vagal innervation. Clinical outcomes in HFrEF trials demonstrate safety and tolerability, with secondary benefits including LVEF increases of approximately 5-8% at 6-12 months and improvements in NYHA class and quality of life, though primary endpoints like mortality or hospitalization reduction have not consistently met significance. For instance, the INOVATE-HF trial reported no difference in the composite of death or worsening heart failure (HR 1.14, p=0.37) but noted enhanced 6-minute walk distance and Minnesota Living with Heart Failure scores. In AF studies, low-level tragus VNS reduced AF burden by up to 85% in short-term applications and lowered inflammatory markers like TNF-α, potentially decreasing recurrence risk. Overall, VNS remains investigational for these indications, with ongoing research refining protocols for broader efficacy.

Stroke rehabilitation

Vagus nerve stimulation (VNS) serves as an adjunctive therapy to rehabilitation for enhancing motor recovery in the upper extremities following ischemic stroke, particularly targeting persistent arm weakness that limits daily function. The U.S. Food and Drug Administration (FDA) granted Breakthrough Device designation and approval in August 2021 for the Vivistim Paired VNS System, enabling its use in patients with moderate to severe upper limb motor deficits to improve arm movement and independence when combined with physical therapy. This approval was based on pivotal randomized controlled trials demonstrating significant gains in motor impairment and function compared to rehabilitation alone. Patient selection focuses on adults with chronic ischemic stroke, typically those at least six months post-event (with studies including up to 10 years), who exhibit unilateral upper extremity weakness without active wrist or thumb extension in the affected arm. Eligibility often requires a (FMA-UE) score between 20 and 50, indicating moderate impairment, alongside exclusion of hemorrhagic stroke, severe comorbidities like depression, or contraindications to surgery. This criteria ensures the therapy targets individuals likely to benefit from enhanced neuroplasticity without excessive risk, as validated in clinical trials where baseline deficits predicted response magnitude. Treatment protocols involve pairing invasive VNS with intensive upper limb rehabilitation over six weeks, consisting of three 90- to 120-minute in-clinic sessions per week, followed by daily at-home exercises. During sessions, brief VNS bursts (0.5 seconds at 30 Hz, up to 0.8 mA intensity, 100 μs pulse width) are precisely timed to coincide with targeted movements, delivered every 5 to 10 seconds to reinforce neural activity. This timing—approximately 300 to 400 stimulation pairs per session—aims to amplify motor learning, with post-protocol assessments showing average FMA-UE improvements of 5 to 9 points sustained at one year. The unique mechanism of paired VNS lies in its ability to enhance cortical plasticity through vagus nerve-mediated release of norepinephrine from the locus coeruleus, which broadens the temporal window for synaptic strengthening during rehabilitation and promotes distributed neural reorganization. This neuromodulatory effect, distinct from standard therapy, facilitates greater task-specific gains by conditioning brainstem nuclei to support long-term motor map changes post-stroke. Recent studies from 2021 to 2025 have confirmed these benefits extend to two- and three-year follow-ups, with ongoing improvements in activity limitations.

Obesity

Vagal blocking therapy using the VBLOC Maestro Rechargeable System received FDA approval in January 2015 as an adjunctive treatment for chronic weight management in adults with obesity. This implantable device delivers high-frequency, low-energy electrical pulses to intermittently block vagal nerve signals between the brain and stomach, promoting satiety and reducing caloric intake without stimulating the nerve directly. Patient selection targets adults aged 18 years and older with a body mass index (BMI) of 40 to 45 kg/m², or 35 to 39.9 kg/m² in the presence of at least one obesity-related comorbidity (e.g., , , or ), who have failed at least one prior supervised weight management program within the past five years. Contraindications include conditions like , large , or need for frequent , as well as patients unable to commit to long-term follow-up. Treatment involves laparoscopic implantation of a neuroregulator in the abdomen and leads around the anterior and posterior near the stomach. Post-implantation, therapy delivers intermittent blocking pulses (5-8 mA, 5000 Hz) for 10-14 hours per day during waking periods, programmable via external controller, combined with lifestyle modifications including diet and exercise. The device battery lasts approximately 5 years, with recharging required weekly. Clinical outcomes from the pivotal ReCharge trial showed 24.4% excess weight loss at 12 months in the active group versus 15.9% in sham (p<0.05), with 52.5% of patients achieving at least 20% excess weight loss; benefits were sustained but modest at 18 months (28.8% vs. 18.7%). Common side effects include implant site pain and gastrointestinal discomfort, with low rates of serious complications (3.7% at 12 months). This therapy offers a reversible neuromodulation option for obesity refractory to behavioral interventions.

Rheumatoid arthritis

Vagus nerve stimulation (VNS) using the SetPoint System received FDA approval on July 30, 2025, as an adjunctive treatment for moderately to severely active rheumatoid arthritis (RA) in adults who have had an inadequate response, loss of response, or intolerance to one or more biologic or targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs). This implantable device activates the cholinergic anti-inflammatory pathway to reduce systemic inflammation and cytokine production, providing a non-pharmacologic option to complement or reduce reliance on immunosuppressive therapies. Patient selection focuses on adults with confirmed RA diagnosis, active disease (e.g., Disease Activity Score 28 [DAS28] >3.2), and documented failure or intolerance to at least one b/tsDMARD such as TNF inhibitors. Candidates undergo rheumatologic evaluation to exclude active infections or contraindications like vagotomy history or cardiac pacemakers. The treatment protocol involves surgical implantation of a in the chest with a lead to the left cervical under . Daily automated stimulation consists of one 1-minute burst (parameters: 1-5 mA, 250 μs pulse width, 10-20 Hz) during sleep or a programmed time, lasting up to 10 years with . It integrates with ongoing RA management, allowing adjustments to DMARDs based on response. Approval was based on the randomized, double-blind RESET-RA (n=242), which demonstrated significant improvements in clinical response rates, with approximately 75% of active patients achieving symptom relief and high satisfaction (78%) at 12 months; 98% persisted with , and serious adverse events were low (1.7%). Outcomes include reduced DAS28 scores and proinflammatory markers like TNF-α, positioning VNS as a targeted immunomodulator for refractory RA as of 2025.

Mechanisms of Action

Central nervous system modulation

Vagus nerve stimulation (VNS) modulates activity primarily through afferent projections from the to key nuclei, influencing widespread neural networks involved in , regulation, and control. The primary site of initial integration is the nucleus tractus solitarius (NTS) in the , which receives approximately 80-90% of vagal afferents and relays signals to higher structures such as the , , and limbic regions. This central modulation is distinct from peripheral effects and contributes to therapeutic outcomes in conditions like by altering excitability and plasticity. A core mechanism of VNS involves cortical desynchronization, which disrupts hypersynchronous neural activity, particularly in epileptic seizures. This effect is mediated by activation of the , a noradrenergic nucleus in the , leading to increased norepinephrine release across cortical and subcortical regions. Norepinephrine elevation enhances arousal and inhibits excessive neuronal firing, with studies showing that lesions abolish VNS's properties. Functional MRI (fMRI) evidence further demonstrates VNS-induced activation of the and insula, correlating with reduced cortical synchrony and improved emotional processing. Recent research as of 2024 also indicates VNS enhances release in the and via projections, supporting cognitive modulation. VNS also induces neurotransmitter changes in limbic structures, enhancing inhibitory transmission and activity to modulate excitability and mood. Projections from the NTS to the promote serotonin release, while indirect pathways influence the and , fostering without direct vagal innervation. For instance, VNS increases serotonin levels in and augments GABA-mediated inhibition in structures, contributing to suppression and effects. Central modulation by VNS promotes bidirectional , particularly through (LTP) in memory-related circuits. Paired with behavioral tasks, VNS strengthens LTP in the and , enabling reorganization of neural networks for learning and . This plasticity is norepinephrine-dependent and supports applications in by enhancing adaptive changes in seizure-prone areas.

Anti-inflammatory pathways

Vagus nerve stimulation (VNS) activates the anti-inflammatory pathway (CAP), an efferent that modulates by inhibiting pro-inflammatory production. This pathway involves electrical impulses traveling along the to the , where they onto the splenic nerve, prompting the release of norepinephrine from splenic nerve terminals. The norepinephrine binds to β2-adrenergic receptors on cholinergic T cells in the , inducing these cells to secrete , which then interacts with α7 nicotinic receptors (α7nAChRs) on macrophages to suppress the synthesis and release of tumor factor-alpha (TNF-α) and other s. In systemic inflammatory conditions, CAP activation via VNS has demonstrated therapeutic potential in animal models by reducing levels of interleukin-6 (IL-6) and (CRP), leading to attenuated and disease severity. For instance, in rheumatoid arthritis (RA) patients, some implantable VNS trials have shown reductions in serum TNF-α and IL-6, correlating with clinical improvements in disease activity scores, though a 2023 meta-analysis of studies found inconsistent evidence overall due to heterogeneity. In sepsis models, VNS protects against lethal endotoxemia by dampening the , with efferent vagal signaling preventing excessive TNF-α release and organ damage. Low-level VNS (LL-VNS), a non-invasive approach using transcutaneous auricular stimulation at subthreshold intensities, modulates chronic by engaging the without requiring surgical implantation, offering a safer option for long-term management of autoimmune disorders. Key evidence from animal models supports these effects, where VNS reduced splenic TNF-α by up to 94% and systemic cytokines by approximately 70% during inflammatory challenges, highlighting the pathway's potency in suppressing immune overactivation. Recent 2024-2025 studies further emphasize VNS's role in modulating microglial toward M2 states via α7nAChR, reducing . This anti-inflammatory mechanism may also contribute to VNS benefits in by lowering peripheral levels that influence mood, as suggested by pilot studies showing modulation.

Autonomic and gut-brain effects

Vagus nerve stimulation (VNS) enhances parasympathetic tone by activating efferent vagal fibers, which predominantly innervate visceral organs and promote restorative physiological responses. This activation increases (HRV), particularly the high-frequency component, serving as a marker of vagal outflow and autonomic balance. Additionally, VNS improves sensitivity, enabling more effective regulation through heightened cardio-vagal responses. In the context of the gut-brain axis, VNS modulates bidirectional signaling via vagal afferents that sense gut microbiota-derived signals and efferents that influence enteric function. These afferents detect microbial metabolites and relay information to the , while efferent stimulation regulates gut motility by enhancing and secretory activity. VNS also reduces by reinforcing tight junctions in the gut barrier, mitigating leaky gut conditions associated with . Furthermore, vagal efferents interact with enterochromaffin cells to modulate serotonin production, which coordinates local gut motility and afferent signaling to the brain. Indirectly, VNS reduces stress responses by inhibiting hypothalamic-pituitary-adrenal (HPA) axis hyperactivity, lowering cortisol release and promoting emotional resilience. This mechanism holds potential for irritable bowel syndrome (IBS) management, where VNS alleviates symptoms through normalized gut signaling and reduced visceral hypersensitivity. Central to these effects are vagal feedback loops that maintain homeostasis, integrating peripheral sensory inputs with central regulatory outputs to balance autonomic and enteric functions.

Devices and Procedures

Invasive devices

Invasive vagus nerve stimulation (VNS) systems consist of surgically implanted components designed for long-term, direct electrical modulation of the vagus nerve. The primary elements include a pulse generator, typically implanted subcutaneously in the left chest wall, and a bipolar lead electrode that wraps helically around the left cervical vagus nerve. The pulse generator is a multiprogrammable device encased in a hermetically sealed titanium housing, powered by a non-rechargeable lithium carbon monofluoride battery with a nominal voltage of 3.3 V and minimal self-discharge rate of less than 1% per year. The lead features two helical electrodes—one anodal and one cathodal—constructed from biocompatible alloys such as platinum-iridium, connected via a silicone-insulated wire to the generator, ensuring targeted stimulation while minimizing tissue damage. The most widely adopted invasive VNS system is the VNS Therapy device, originally developed by Cyberonics and now manufactured by following the company's acquisition in 2016. Early models, such as the NCP Model 100 approved by the FDA in 1997, delivered pulses with programmable parameters including output current ranging from 0.25 to 3.5 mA, of 130–750 μs, and of 1–30 Hz. Battery life for these systems typically spans 5–10 years, depending on stimulation settings and impedance (e.g., approximately 3 kΩ), after which the generator requires surgical replacement. Over 125,000 such devices have been implanted globally for and other indications, as of 2023, demonstrating the system's reliability in chronic use. Advancements in invasive VNS technology have focused on enhancing compatibility and longevity while maintaining therapeutic efficacy. Modern iterations, such as LivaNova's Model 106 (AspireSR), Model 1000 (SenTiva, introduced in 2017), and Model 1000-D (SenTiva DUO, launched in 2023 with dual-pin compatibility for lead upgrades), incorporate full-body MRI-conditional features up to 3.0 , allowing safe imaging without device removal—a critical for patients requiring frequent diagnostics. These models also integrate advanced sensing capabilities, like automatic stimulation triggered by detection, to optimize control. Despite these innovations, technology remains non-rechargeable, with projected lifespans extending to over 10 years in low-duty-cycle applications; experimental rechargeable or batteryless prototypes are under investigation but not yet standard in approved systems. The historical evolution of invasive VNS devices traces back to early prototypes in the , pioneered by researchers at Cyberonics based on foundational demonstrating antiseizure effects. The first human occurred in 1988, marking the transition from experimental setups to engineered implants with refined electrode designs. Subsequent FDA approvals in 1997 for , followed by humanitarian device exemptions and expansions, drove iterative improvements across ten generator models by the , shifting from basic pulse delivery to sophisticated, patient-adaptive systems. This progression reflects a balance between , , and clinical performance demands.

Non-invasive devices

Non-invasive vagus nerve stimulation (nVNS) devices deliver electrical impulses to the through the skin without requiring surgical implantation, offering a reversible and accessible alternative to invasive methods. These devices primarily target peripheral branches of the using surface electrodes, enabling patient self-administration at home. By applying lower-intensity currents compared to implanted systems, nVNS modulates autonomic functions and activity with reduced risk of procedural complications. Two main categories of nVNS devices exist: transcutaneous auricular vagus nerve stimulation (taVNS) and transcutaneous cervical vagus nerve stimulation (tcVNS). taVNS devices stimulate the auricular branch of the via electrodes placed on the , typically the tragus or cymba concha, to activate afferent fibers that project to brainstem nuclei like the nucleus tractus solitarii. Examples include the Nurosym device by Parasym, a wearable clip system that delivers targeted pulses for conditions such as , and the earlier NEMOS system, which received in for treatment. These auricular stimulators operate at currents of 0.5–10 mA, adjusted to perceptual thresholds, for sessions lasting 1–4 hours multiple times daily. In contrast, tcVNS devices apply stimulation to the cervical branch of the by positioning electrodes on the neck over the . The gammaCore, developed by electroCore, is a prominent handheld example that uses bipolar electrodes to deliver 25 Hz pulses at intensities up to 60 mA for 90–120 second cycles, targeting pain pathways. The U.S. (FDA) cleared gammaCore via the pathway in 2015 for episodic and expanded approval in 2017 for acute treatment in adults, with further extensions to adolescents and preventive use. Both taVNS and tcVNS devices emphasize portability and user control, allowing on-demand or scheduled use without medical supervision after initial training. However, their shallower tissue penetration limits direct activation of the main vagal , potentially leading to variable across individuals and conditions compared to invasive approaches. Common limitations include inconsistent stimulation parameters across studies and devices, as well as milder but transient side effects like skin or discomfort. For instance, while gammaCore has demonstrated pain relief in disorders, broader applications remain under investigation due to these variability factors.

Implantation and stimulation protocols

The implantation of a stimulator (VNS) is typically performed as an outpatient surgical procedure under general , with the patient positioned and the extended to facilitate access. A transverse incision, approximately 2-3 cm in length, is made on the left side of the at the level of the to expose the left within the ; a second incision, 2.5-3 cm long, is created in the left upper chest, about 5 cm below the , to form a subcutaneous pocket for the . The is carefully dissected over a length of about 4 cm, and helical s are wrapped around it using a triple-helix configuration—starting with the central coil, followed by the upper and lower coils—before securing the assembly with a strain relief suture or anchor to accommodate movement. The lead is then tunneled subcutaneously to the chest incision, where the is implanted into the pocket and fixed with non-absorbable sutures. The entire procedure generally lasts 1 to 2 hours, with closure using absorbable sutures subcutaneously and non-absorbable sutures for the skin. Potential risks include , which occurs in approximately 2-3% of cases, as well as hoarseness or . Programming of the VNS device begins 2 weeks post-implantation to permit and is conducted by a neurologist or trained using a handheld programming that communicates wirelessly with the . Initial settings are conservative to minimize side effects, typically including an output current of 0.25 mA, a of 30 Hz, a of 500 μs, and a of 30 seconds on followed by 5 minutes off. involves incremental adjustments, often monthly, increasing the output current in steps of 0.25 mA (up to 1.25-1.75 mA) and potentially shortening the off time while monitoring for tolerability, with parameters fine-tuned based on clinical response and patient feedback. Standard stimulation protocols employ a fixed duty cycle of 30 seconds of every 5 minutes to efficacy and , though adaptive options allow patients or caregivers to activate extra via a swipe, triggering a 60-second burst at 0.25-0.5 mA higher than the baseline current. Monitoring during and ongoing use may incorporate to assess cardiac effects, voice recordings to detect laryngeal changes, or patient diaries tracking symptoms like or discomfort. Post-operative care emphasizes wound management, with assessment for signs of and removal of dressings prior to device activation at the 2-week follow-up. Patients receive on magnet use and activity restrictions, while regular clinic visits include device interrogation via the programming to verify battery status, review stimulation history, and optimize settings as needed.

Safety Profile

Surgical complications

Surgical complications associated with vagus nerve stimulation (VNS) implantation primarily arise during the intraoperative phase or shortly postoperatively, with overall rates for primary implantation reported at approximately 8.6% to 13.4% across large cohorts. These risks are generally low due to standardized surgical techniques, but they necessitate careful patient selection and perioperative management to minimize adverse outcomes. Intraoperative complications include potential nerve damage, such as unilateral , which occurs in 1% to 2.7% of cases and may result from direct manipulation of the during electrode placement. Bleeding, manifesting as peritracheal hematoma, affects about 1.9% of patients and can require emergent intervention if it compromises airway patency. General anesthesia-related issues, such as respiratory compromise, are rare but possible, aligning with broader neurosurgical risks during and tunneling. Postoperative complications encompass infections at the surgical site, occurring in 2% to 3.5% of implantations, often necessitating device removal if deep-seated. Lead migration or fracture represents another concern, with fracture rates around 3% and migration contributing to hardware revisions in up to 5% of cases over five years. , though less common, can arise from tension on incision sites and occurs in fewer than 2% of procedures based on multicenter data. Management strategies emphasize prophylactic antibiotics, such as a single preoperative dose of , which has been shown to reduce rates to approximately 2%. Revision surgeries are required in about 10% of patients over their lifetime with the device, typically for lead issues or , with higher rates (21.4%) observed in lead-specific revisions compared to primary procedures. These interventions often preserve device functionality while addressing complications promptly.

Stimulation side effects

Vagus nerve stimulation (VNS) commonly induces transient side effects during the active "on" phase of stimulation pulses, primarily due to activation of laryngeal and respiratory branches of the vagus nerve. The most frequent symptoms include voice alteration or hoarseness, affecting approximately 45% of patients, along with cough in about 15% and dyspnea in 14%. These effects typically occur synchronously with stimulation cycles and are generally mild to moderate in severity. Certain side effects are dose-dependent, correlating with stimulation parameters such as output current intensity and pulse width. , reported in around 16% of cases, and , occurring in about 16%, often arise from higher stimulation levels and can be effectively mitigated by reducing the current or adjusting . Rarer complications include , with an incidence of approximately 0.1% during initial testing and even fewer late-onset cases, potentially leading to syncope if severe. risk, though uncommon, has been documented in isolated reports, often linked to impaired coordination during stimulation; patients can activate a to temporarily disable the device and avert such events. Over time, many patients develop tolerance to these stimulation-induced effects, with studies showing a substantial decrease in both frequency and severity—often by half or more—within the first year of as the body adapts.

Long-term risks and management

Long-term use of vagus nerve stimulation (VNS) devices is generally associated with a stable safety profile, as evidenced by multiple studies spanning over a decade of follow-up in patients with refractory and . In a single-center of pediatric patients with a mean follow-up exceeding 10 years, the demonstrated sustained efficacy without emerging safety concerns, with hardware-related interventions accounting for the majority of revisions. Similarly, a 30-year review of VNS data confirmed that adverse events decrease in frequency and severity over time, supporting its role as a lifelong treatment option for many patients. Chronic risks primarily involve device hardware issues, such as depletion necessitating surgical . Batteries in current VNS generators typically last 3 to 10 years, depending on parameters, with procedures comprising nearly half of all VNS-related surgeries in long-term cohorts. around the can develop over years of implantation, potentially leading to reduced or the need for lead revision, though this is manageable and occurs in a minority of cases. Potential cardiac remodeling represents another consideration, as prolonged VNS may influence autonomic balance; however, clinical data indicate it more often mitigates adverse remodeling in models rather than inducing it, with rare reports of late-onset bradyarrhythmias requiring . Ongoing management emphasizes regular monitoring to mitigate these risks, including annual clinical check-ups to assess function and status, alongside impedance testing to detect lead integrity issues early. Quality-of-life assessments, such as standardized scales for control or , are integrated into follow-up protocols to evaluate holistic outcomes. Explantation rates range from 5% to 18% over extended periods, often due to inefficacy, , or preference, with procedures generally low-risk when performed by experienced surgeons. VNS devices are compatible with MRI under specific protocols, requiring deactivation and parameter adjustments to avoid heating or malfunction, while (deep heat therapy) is contraindicated due to risks of damage.

Historical Development

Early physiological studies

In the late 19th century, early explorations into the physiological effects of vagus nerve manipulation laid foundational insights for later stimulation research. American neurologist James L. Corning proposed in the 1880s that stimulating the vagus nerve could reduce cerebral blood flow, potentially alleviating epileptic seizures by decreasing intracranial pressure and calming neural hyperactivity. He developed rudimentary devices, such as a neck collar applying mechanical pressure and electrical impulses to the carotid sheath encompassing the vagus, marking one of the first conceptual links between vagal modulation and central nervous system (CNS) influence. During the and , experimental studies expanded on stimulation's impacts on peripheral organs and the CNS, primarily using animal models. Stimulation was shown to elicit pronounced effects on the heart, inducing and through parasympathetic activation, as demonstrated in canine preparations where electrical pulses slowed cardiac rhythm via release. Similarly, vagal activation influenced pulmonary function, inhibiting respiratory drive and altering inflation reflexes in dogs, highlighting the nerve's role in coordinating cardiorespiratory . Key work by in 1922 examined vagotomy's effects on gastric innervation, revealing reduced secretory activity and , which shifted understanding toward the vagus as a modulator of visceral functions beyond mere reflex pathways. In parallel, investigations into CNS effects revealed inhibitory potentials. In 1938, and Bremer reported that vagus stimulation in synchronized cortical electrical activity, suggesting ascending projections that could dampen neural excitability and produce sedative-like inhibition in the and higher centers. Dog models further corroborated CNS modulation, with 1930s experiments showing stimulation-induced suppression of cortical , akin to a generalized inhibitory response. These findings marked a conceptual from viewing the vagus primarily as a peripheral to recognizing its neuromodulatory capacity, influencing widespread autonomic and encephalic processes and paving the way for therapeutic applications.

Epilepsy treatment origins

The origins of vagus nerve stimulation (VNS) as a treatment for epilepsy trace back to the 1980s, when research shifted from basic physiological investigations to targeted translational studies in animal models. Building on prior work exploring vagal influences on brain activity, Jacob Zabara proposed in 1985 that electrical stimulation of the vagus nerve could interrupt hypersynchronous neural discharges underlying epileptic seizures. In canine experiments, repetitive vagal stimulation effectively abolished or interrupted motor seizures induced by strychnine, demonstrating the potential for peripheral nerve modulation to suppress central epileptic activity without direct brain intervention. This breakthrough inspired the commercialization of VNS technology, with Cyberonics Inc. founded in 1987 to engineer implantable devices for clinical application. The first human implantation occurred in 1988, performed by neurologist J. Kiffen Penry and neurosurgeon William Bell at School of Medicine on a 25-year-old with refractory epilepsy. Initial human trials from 1988 to 1993, including pilot feasibility studies (E01 and E02) involving small cohorts totaling 14 , evaluated intermittent stimulation protocols and reported meaningful seizure reductions, with up to 40% frequency decrease in some individuals at short-term follow-up and responder rates (≥50% reduction) reaching 50% in extended observations of the E02 group. These open-label studies established preliminary safety and tolerability, with adverse effects limited primarily to transient hoarseness and . Building on these results, larger-scale pivotal trials confirmed VNS efficacy for . The E03 trial (1994), a multicenter randomized double-blind study with 114 adults, compared high-frequency stimulation (output current 1.25 times the threshold) to low-frequency (no output current during ON cycles), yielding a 24.5% seizure frequency reduction in the high group versus 6.1% in the low group at three months. The subsequent E05 trial (1995–1996), involving 199 patients, replicated these findings with a 28% reduction in the high-stimulation arm. Cumulatively, these trials across approximately 25 patients in early non-randomized phases and hundreds in controlled settings demonstrated sustained benefits, leading to U.S. approval in July 1997 for VNS as adjunctive therapy in reducing partial-onset s in patients aged 12 years and older to antiepileptic drugs.

Expansion to modern indications

Following the initial approval for epilepsy in 1997, vagus nerve stimulation (VNS) expanded to other indications, beginning with . In 2005, the U.S. (FDA) approved the VNS Therapy System as an adjunctive long-term for chronic or recurrent in adults aged 18 years or older who had not adequately responded to four or more adequate trials. This approval was based on the D-02 randomized, double-blind, controlled acute-phase trial, which demonstrated significant improvements in response rates compared to sham stimulation, with sustained benefits observed in open-label extensions. In , FDA approval for was expanded to include children aged 4 years and older with partial-onset seizures. In the , VNS gained further regulatory milestones for cardiovascular and pain-related conditions. The VITARIA System, an implantable VNS device for autonomic regulation therapy, received CE Mark approval in Europe in for treating patients with moderate to severe chronic ( Heart Association Class II/III) and reduced , aiming to improve heart function through vagal modulation. In , the FDA approved the Rechargeable System, a vagal blocking device, for treatment in adults with between 35 and 45 kg/m² and at least one related comorbid condition. For headache disorders, the gammaCore non-invasive VNS device was cleared by the FDA in for the acute treatment of pain associated with episodic in adult patients, with subsequent of external vagal stimulators for into Class II (special controls). This clearance was supported by randomized trials showing rapid pain relief in a significant proportion of attacks. Entering the 2020s, VNS applications extended to neurological . In August 2021, the FDA approved the Vivistim Paired VNS System as an adjunctive therapy to intensive for improving upper extremity motor deficits in adults with chronic ischemic (at least six months post-event), marking a breakthrough designation for its role in enhancing during therapy. Clinical trials demonstrated that pairing VNS with led to clinically meaningful gains in motor , with improvements sustained beyond the period. For , VNS has received attention through investigational designations and trials exploring its potential to alleviate and via anti-inflammatory and autonomic effects, though full approval remains pending as of 2025. In July 2025, the FDA approved the SetPoint System, an implantable VNS device, for the of moderate to severe in adults who have had an inadequate response to inhibitors. Globally, VNS adoption has grown beyond the U.S., with CE Mark approvals in facilitating broader use for , , and headaches since the mid-2000s. In , regulatory approvals have accelerated, particularly in where implantable VNS for was cleared in 2010, followed by expansions to and off-label applications in , driven by increasing clinical evidence and market demand. has also proliferated, including for inflammatory conditions, contributing to overall growth in VNS implantation rates worldwide, with over 100,000 devices placed by the late .

Ongoing Research

Technological advancements

Recent innovations in vagus nerve stimulation (VNS) have centered on closed-loop systems that enable real-time adaptive . In 2025, researchers developed a fully automated VNS system featuring a biocompatible, miniaturized implant with cuff electrodes that integrates (EEG) for detection, allowing the device to deliver stimulation precisely when abnormalities are detected, thereby enhancing efficacy and reducing unnecessary interventions. This closed-loop approach contrasts with earlier open-loop devices by responding dynamically to physiological signals, potentially improving patient outcomes in management. Advancements in fiber-specific targeting have addressed limitations of non-selective , which can activate unintended fibers and cause side effects. The 2025 Feinstein method, developed by researchers at the Feinstein Institutes, employs intermittent interferential sinusoidal current through multi-contact epineural cuffs to selectively activate A-fibers while minimizing engagement of B- and C-fibers, enabling spatiotemporal control over organ-specific responses. This technique, supported by a $3 million NIH grant, aims to enhance precision and safety by steering electrical currents to target therapeutic fibers without eliciting adverse reactions like hoarseness or coughing. Miniaturization efforts have progressed significantly, with externally powered micro-implants emerging as a key development. In 2024, prototypes of miniature externally powered stimulators, approximately 50 times smaller than conventional VNS devices, were implanted using minimally invasive single-incision procedures, relying on external power sources to eliminate bulky batteries. By 2025, ultrasound-based charging mechanisms further advanced battery life and device longevity, as demonstrated in an ultrasonically powered neuromodulation platform that wirelessly delivers energy through tissue without inductive coils, supporting chronic implantation with enhanced safety via charge balancing. Non-invasive VNS enhancements have expanded accessibility through low-level devices designed for home use. Recent 2025 models of transcutaneous auricular VNS (taVNS) devices, such as ear-clip stimulators, deliver low-intensity pulses at user-controlled settings, allowing daily at-home sessions to modulate autonomic function without surgical intervention. These portable systems, often app-integrated for personalized protocols, represent an evolution from clinical-only applications, promoting broader adoption for and mild therapeutic needs.

Emerging clinical applications

Recent reviews synthesizing evidence from 2021 to 2025 have underscored the potential of vagus nerve stimulation (VNS) paired with to enhance motor recovery in patients, particularly for upper extremity function. In the VNS-REHAB trial, a pivotal randomized controlled study, participants receiving active implantable VNS during intensive demonstrated a mean improvement of 5.0 points on the Fugl-Meyer Assessment-Upper Extremity (FMA-UE) scale compared to 2.4 points in the control group, with 47% achieving clinically meaningful gains of at least 6 points versus 24% in controls. Extensions of this trial, including one-year follow-up data from 74 participants published in 2025, revealed sustained benefits, with 66.2% maintaining clinically meaningful responses in motor impairment, activity levels, and . Case reports within these reviews have documented notable motor gains, such as up to 28-point increases on the FMA-UE scale in individual patients, aligning with overall trends of 20-30% functional improvements in targeted metrics. Investigational applications of VNS in (TRD) have advanced with large-scale trials providing robust evidence of symptom relief. The RECOVER trial, a multicenter randomized controlled study published in 2025 involving 493 adults with markedly TRD, demonstrated that adjunctive VNS therapy over 12 months led to significant reductions in depressive symptoms across multiple , including the Montgomery-Åsberg Scale (MADRS), with large benefits observed in remission, response, and partial response rates compared to treatment-as-usual alone. Long-term analyses from this cohort indicated durable effects, with approximately 40% of participants achieving remission in severe cases, highlighting VNS as a viable option for patients unresponsive to multiple prior interventions.01390-1/fulltext) Beyond these areas, emerging data support VNS in other conditions. A 2025 retrospective study of 40 adults with drug-resistant reported improvements in seizure frequency, duration, and intensity in 65% of patients over a median follow-up of more than five years, alongside enhancements in quality-of-life measures related to health status. For chronic , clinical trials and meta-analyses from 2024-2025 indicate that transcutaneous VNS reduces pain intensity, though efficacy is more pronounced in headache-related pain than in purely neuropathic types, with observational studies showing up to 87% pain reduction in combined approaches. Potential roles in , particularly for persistent and symptoms like , have been explored in pilot studies up to 2024, but evidence remains preliminary and has been de-emphasized following mixed outcomes post-2023, with transcutaneous VNS showing modest improvements in dyspnea and energy levels in small female cohorts. Non-invasive VNS modalities are addressing gaps in applications for general and immune modulation, offering accessible alternatives without surgical risks. Studies from 2024-2025 demonstrate that transcutaneous auricular VNS activates the pathway, reducing pro-inflammatory cytokines in autoimmune conditions like and , with adjunctive benefits in symptom alleviation and overall autonomic balance. These approaches also promote by enhancing and exercise capacity in chronic illness populations, positioning non-invasive VNS as a promising tool for broad immunomodulatory effects.

References

  1. [1]
    Neuroanatomy, Cranial Nerve 10 (Vagus Nerve) - StatPearls - NCBI
    The vagus nerve (cranial nerve [CN] X) is the longest in the body, containing both motor and sensory functions in afferent and efferent regards.
  2. [2]
    Vagus nerve stimulation - Mayo Clinic
    Dec 20, 2024 · Vagus nerve stimulation involves using a device to send electrical impulses to the vagus nerve. The vagus nerve is the main nerve of the system ...
  3. [3]
    Vagus Nerve Stimulator - StatPearls - NCBI Bookshelf
    VNS is a Food and Drug Administration (FDA) approved treatment for various conditions like chronic epilepsy, refractory epilepsy, and depression.
  4. [4]
    Vagus nerve stimulation (VNS): recent advances and future directions
    Oct 4, 2024 · Vagus nerve stimulation (VNS) is emerging as a unique and potent intervention, particularly within neurology and psychiatry.Mechanisms Of Vns · Vns For Epilepsy · Vns For Depression
  5. [5]
    Vagus Nerve as Modulator of the Brain–Gut Axis in Psychiatric and ...
    Mar 13, 2018 · The vagus nerve is responsible for the regulation of internal organ functions, such as digestion, heart rate, and respiratory rate, as well as ...
  6. [6]
    Organ- and function-specific anatomical organization of vagal fibers ...
    Vagal fibers travel inside fascicles and form branches to innervate organs and regulate organ functions. Existing vagus nerve stimulation (VNS) therapies ...
  7. [7]
    The Vagus Nerve in the Neuro-Immune Axis - PubMed Central
    Nov 2, 2017 · The VN is composed of 80% afferent fibers conveying taste, visceral and somatic information and 20% efferent fibers involved in the control of ...
  8. [8]
    Heart Rate Variability and Cardiac Vagal Tone in ... - PubMed Central
    Feb 20, 2017 · More specifically, HF corresponds to vagal tone when between 0.15 and 0.40 Hz, which means between 9 and 24 cycles per minute regarding ...
  9. [9]
    The polyvagal theory: phylogenetic substrates of a social nervous ...
    The evolution of the autonomic nervous system provides an organizing principle to interpret the adaptive significance of physiological responses in ...
  10. [10]
    A Review of Parameter Settings for Invasive and Non ... - Frontiers
    A Review of Parameter Settings for Invasive and Non-invasive Vagus Nerve Stimulation (VNS) Applied in Neurological and Psychiatric Disorders.
  11. [11]
    A Review of Parameter Settings for Invasive and Non-invasive ...
    Jul 13, 2021 · Uthman et al. (1993) used slightly different VNS parameters (500 μs pulses, 50 Hz, up to 120 s ON, 5–20 min OFF, 20 weeks duration with no ...
  12. [12]
    Specific vagus nerve stimulation parameters alter serum cytokine ...
    Apr 10, 2020 · This application of vagus nerve stimulation activates the inflammatory reflex, reducing levels of inflammatory cytokines during inflammation.Missing: biophysical | Show results with:biophysical
  13. [13]
    Vagus Nerve Stimulation - PMC - PubMed Central
    Vagus nerve stimulation (VNS) refers to any technique that stimulates the vagus nerve, including manual or electrical stimulation.Vagus Nerve Stimulation · Methods Of Vagus Nerve... · Studies Of Vns For...
  14. [14]
    Data Driven Control of Vagus Nerve Stimulation for the ... - Frontiers
    VNS involves sending electrical pulses through a cuff electrode to the vagus nerve, with the electrical pulse characterized by several parameters such as pulse ...
  15. [15]
    Vagus nerve stimulation: Invasive or noninvasive? - PubMed Central
    Emerging noninvasive transcutaneous VNS (tVNS) approaches are not limited to treatments of migraine and headache.
  16. [16]
    Comparison of traditional and closed loop vagus nerve stimulation ...
    Closed loop VNS trends towards a higher rate of VNS response after two years of treatment, especially among generalized epilepsy patients.
  17. [17]
    Vagus nerve stimulation activates nucleus of solitary tract neurons ...
    Oct 22, 2021 · Our chief findings indicate that the KC reduced basal spontaneous activity of second-order NTS neurons receiving myelinated vagal input by 85%.
  18. [18]
    P970003 - Premarket Approval (PMA) - FDA
    01/27/1997 ... Approval for the NeuroCybernetic Prosthesis (NCP(R)) System which includes the Model 100 NCP Generator, the Model 300 Series NCP Vagus Nerve ...
  19. [19]
    Evolution of the Vagus Nerve Stimulation (VNS) Therapy System ...
    The VNS Therapy received FDA approval in 1997 for use as an adjunctive therapy in reducing the frequency of partial onset seizures which are refractory to ...
  20. [20]
    [PDF] SUMMARY OF SAFETY AND EFFECTIVENESS DATA (SSED)
    The primary objective of the study was to demonstrate that stimulation of the vagus nerve reduces the frequency of partial onset seizures. Two weeks after ...
  21. [21]
    Vagus Nerve Stimulation - Medical Clinical Policy Bulletins - Aetna
    VNS with a miniaturized neurostimulator was safe and well-tolerated and reduced signs and symptoms of RA in patients with multidrug-refractory disease.
  22. [22]
    Clinical Benefit of Vagus Nerve Stimulation for Epilepsy
    Inclusion criteria consisted of patients ≥12 years old with medically intractable seizures, defined as a frequency of ≥6 per month and predominantly partial ...
  23. [23]
    [PDF] Vagus and External Trigeminal Nerve Stimulation - UHCprovider.com
    May 1, 2025 · Medically refractory epileptic seizures with failure of two or more trials of single or combination antiepileptic drug therapy or intolerable ...
  24. [24]
    Programming in Vagus nerve stimulation therapy: Consensus from a ...
    According to Delphi panel, VNS should be started at 0.25 mA and for further titration, VNS should usually be increased by 0.25 mA steps either by an epilepsy ...
  25. [25]
    Vagus Nerve Stimulation (VNS): What It Is, Uses & Side Effects
    The use of VNS is limited to a select group of individuals who have treatment-resistant epilepsy or treatment-resistant depression. More recently, VNS has been ...
  26. [26]
    Rates and Predictors of Seizure Freedom With Vagus Nerve ... - NIH
    These studies show that about 50% to 60% of patients achieve ≥50% reduction in seizure frequency after 2 years of treatment, and response rates increase over ...
  27. [27]
    Predictive factors for successful vagus nerve stimulation in patients ...
    Jul 26, 2023 · This retrospective study evaluated vagus nerve stimulation (VNS) as an adjuvant therapy for patients with drug-resistant epilepsy from six epilepsy centers in ...Abstract · Introduction · Results · Discussion
  28. [28]
    [PDF] Summary of S~fety and Effectiveness Data - accessdata.fda.gov
    Jul 15, 2005 · The VNS Therapy System is indicated for the adjunctive long-term treatment of chronic or recurrent depression for patients 18 years of age or ...
  29. [29]
    Comparative Effectiveness of Transcutaneous Auricular Vagus ...
    The primary outcome was the 17-item Hamilton Depression Rating Scale (HAM-D17) measured every two weeks by trained interviewers blinded to the treatment ...Missing: HAMD- | Show results with:HAMD-
  30. [30]
    VNS Therapy in Treatment-Resistant Depression: Clinical Evidence ...
    Apr 19, 2006 · ... approved by the US Food and Drug Administration for treatment-resistant depression in July, 2005. The physiology of the vagus nerve, mechanics ...
  31. [31]
    A prospective, multi-center randomized, controlled, blinded trial of ...
    Few treatment options exist for patients with difficult-to-treat depression (DTD). One potentially efficacious treatment is vagus nerve stimulation (VNS): ...
  32. [32]
    From antidepressants and psychotherapy to oxytocin, vagus nerve ...
    Vagus nerve stimulation (VNS) offers an alternative approach to treating depression ... Combining pharmacotherapy and psychotherapy or monotherapy for ...
  33. [33]
    A Comprehensive Review of Vagus Nerve Stimulation for Depression
    Response rates increased from 37% at 3 months to 53% at 12 months and remission rates increased from 17% at 3 months to 33% at 12 months. The time course of VNS ...The Invention Of Vns · Noninvasive Vns · Concurrent Vns/fmri
  34. [34]
    Vagus Nerve Stimulation, Depression, and Inflammation - Nature
    Aug 14, 2007 · Acetylcholine, the principal vagus neurotransmitter, inhibits the production of proinflammatory cytokines through a mechanism dependent on the ...
  35. [35]
    [PDF] De Novo Summary (DEN150048) Page 1 of 22 gammaCore Non ...
    The safety and effectiveness of the gammaCore Non-invasive Vagus Nerve Stimulator has not been established in the acute treatment of chronic cluster headache.
  36. [36]
    gammaCore® Receives FDA Clearance for the Acute Treatment of ...
    Feb 1, 2018 · gammaCore receives FDA clearance for the acute treatment of pain associated with migraine headache in adult patients.
  37. [37]
    [PDF] Summary - 510(k) Premarket Notification - FDA
    Nov 27, 2018 · gammaCore is indicated for: • Adjunctive use for the preventive treatment of cluster headache in adult patients. • The acute treatment of pain ...
  38. [38]
    Noninvasive vagus nerve stimulation as acute therapy for migraine
    Jul 24, 2018 · Noninvasive vagus nerve stimulation as acute therapy for migraine: The randomized PRESTO ... pain or being pain-free 2 hours poststimulation ( ...
  39. [39]
    [PDF] Instructions for Use for gammaCore SapphireTM SLC
    gammaCore™ provides a mild electrical stimulation to the vagus nerve, which runs through the neck and carries information to the central nervous system. Each ...Missing: protocol dosing
  40. [40]
    [PDF] February 12, 2021 Electrocore, Inc. Mike Romaniw VP, Quality ...
    Feb 12, 2021 · the gammaCore Sapphire device cleared in K191830. No changes in ... gammaCore Sapphire for the preventive treatment of migraine headache.
  41. [41]
    gammaCore Dosing Information
    Using gammaCore to prevent migraine headache. Daily: Give yourself 2 treatments (morning and night) consisting of 2 consecutive two-minute stimulations.Missing: contraindications | Show results with:contraindications
  42. [42]
    Vagus Nerve Stimulation for Treating Adults With Severe Fibromyalgia
    Although VNS has been FDA approved for some forms of epilepsy and depression, it has yet to receive FDA approval for FMS.
  43. [43]
    Molecular Aspects of Regional Pain Syndrome - PubMed Central - NIH
    For instance, vagal nerve stimulation might have the potential for treating CRPS through the cholinergic anti-inflammatory pathway. 1. Introduction. Complex ...
  44. [44]
    Evaluating the efficacy and acceptability of vagus nerve stimulation ...
    Feb 21, 2024 · Recently, vagus nerve stimulation (VNS) has emerged as a promising therapy for fibromyalgia. Nonetheless, despite the increasing number of ...
  45. [45]
    Evaluating the efficacy and acceptability of vagus nerve stimulation ...
    Feb 20, 2024 · As the efficacy of VNS for fibromyalgia is undetermined, the intervention group's eligibility criteria will be limited to the use of VNS alone.Missing: breakthrough | Show results with:breakthrough
  46. [46]
    Transcutaneous Vagus Nerve Stimulation on Fibromyalgia- Double ...
    Vagus nerve stimulation is a FDA-approved treatment for different pathologies. The regulation of the autonomic and immune systems and a specific effect on ...
  47. [47]
    NCT07080749 | The Effect of Vagus Nerve Stimulation on Pain and ...
    Jul 23, 2025 · The goal of this clinical trial is to examine the effect of vagus nerve stimulation (VNS) on chronic, widespread pain in adults.
  48. [48]
    Role of Vagus Nerve Stimulation in the Treatment of Chronic Pain
    [44] found that the pain relief rate (≥50% reduction in visual analog scale [VAS] score) was 64.6% and 39.6% of patients achieved pain-free status at 2 h after ...
  49. [49]
    Transcutaneous vagus nerve stimulation effects on chronic pain
    This meta-analysis indicates that tVNS shows promise as an effective intervention for managing pain intensity in chronic pain conditions.
  50. [50]
    Vagus Nerve Stimulation and Inflammation in Cardiovascular Disease
    Sep 18, 2023 · In the setting of heart failure, VNS was found to improve systolic function and reverse cardiac remodeling. In summary, existing experimental ...
  51. [51]
    Vagus Nerve Stimulation for the Treatment of Heart Failure - JACC
    The results of the study show that VNS did not improve the risk of death or HF events among patients with HF and a reduced LVEF. Perspectives. COMPETENCY IN ...
  52. [52]
    Vagus Nerve Stimulation and Atrial Fibrillation: Revealing the Paradox
    LLVNS is a promising novel therapeutic modality to treat AF and further research will further elucidate the underlying anti-arrhythmogenic mechanisms.
  53. [53]
    Role of the Autonomic Nervous System in Atrial Fibrillation
    Apr 25, 2014 · Chronic vagus nerve stimulation improves autonomic control and attenuates systemic inflammation and heart failure progression in a canine ...Cardiac Autonomic... · Autonomic Effects On... · Vagal Nerve Stimulation
  54. [54]
    TREAT AF (Transcutaneous Electrical Vagus Nerve Stimulation to ...
    Jan 29, 2020 · Noninvasive neuromodulation using low-level tragus stimulation significantly decreased atrial fibrillation burden and decreased tumor necrosis ...
  55. [55]
    [PDF] SUMMARY OF SAFETY AND EFFECTIVENESS DATA (SSED)
    The Vivistim® System when used as intended, provides a drug-free way to treat upper extremity motor deficits associated with a stroke by pairing rehabilitation ...
  56. [56]
    An Exploratory Study of Predictors of Response to Vagus Nerve ...
    Nov 4, 2019 · VNS-treated participants had greater improvement in FMA-UE score at day-1 post therapy (8.63 ± 5.02 points) versus control (3.79 ± 5.04 points; ...Methods · Brain Mri Acquisition And... · Vns And Control Responders
  57. [57]
    Long-Term Outcomes of Vagus Nerve Stimulation Paired With ...
    May 7, 2025 · At 1 year, compared with baseline, there were improvements in impairment (Fugl-Meyer Assessment UE, 5.23 [95% CI, 4.08–6.39]; P<0.001) activity ...
  58. [58]
    Norepinephrine and serotonin are required for vagus nerve ...
    We tested the hypothesis that cortical norepinephrine and serotonin are required for VNS-dependent enhancement of motor cortex plasticity.
  59. [59]
    Targeted Vagus Nerve Stimulation for Rehabilitation After Stroke
    Mar 28, 2019 · One such targeted strategy uses brief bursts of Vagus Nerve Stimulation (VNS) paired with rehabilitation to enhance plasticity and support recovery of upper ...
  60. [60]
    The vagus afferent network
    Recent work has also suggested that VNS may reorga- nize functional circuits within the limbic system. Using. BOLD fMRI data in rats receiving VNS, Cao et al.Missing: serotonin | Show results with:serotonin
  61. [61]
  62. [62]
  63. [63]
    Mechanism and Applications of Vagus Nerve Stimulation - PMC
    Feb 14, 2025 · VNS exhibits anti-inflammatory effects, modulates neurotransmitter release, enhances neural plasticity, inhibits apoptosis and autophagy.<|control11|><|separator|>
  64. [64]
  65. [65]
    Vagus Nerve Stimulation (VNS) Modulates Synaptic Plasticity in the ...
    Jun 5, 2024 · Our study suggests that VNS consolidates the extinction of drug-seeking behavior by reversing drug-induced changes in synaptic AMPA receptors in the IL.
  66. [66]
    Splenic nerve is required for cholinergic antiinflammatory pathway ...
    We show that vagus nerve stimulation during endotoxemia specifically attenuates TNF production by spleen macrophages in the red pulp and the marginal zone.
  67. [67]
    Splenectomy inactivates the cholinergic antiinflammatory pathway ...
    Jun 19, 2006 · Vagus nerve stimulation significantly reduces TNF levels in the spleen (94%) and liver (40%), but not in the lung (20%) (Fig. 1 A).
  68. [68]
    The cholinergic anti-inflammatory pathway inhibits inflammation ...
    Apr 13, 2023 · A direct binding of NA released by splenic nerve to β2-AR on macrophages might explain the anti-inflammatory effect induced by vagus or splenic ...
  69. [69]
    Vagus nerve stimulation inhibits cytokine production and attenuates ...
    Jul 5, 2016 · Vagus nerve stimulation inhibits cytokine production and attenuates disease severity in rheumatoid arthritis ... Third, we observed reduced TNF ...
  70. [70]
    Non-invasive vagus nerve stimulation in anti-inflammatory therapy
    Vagus nerve stimulation inhibits cortical spreading depression via glutamate-dependent TrkB activation mechanism in the nucleus tractus solitarius.Introduction · Mechanisms of vagus nerve... · Expanded clinical applications...
  71. [71]
    Vagus Nerve Stimulation and the Cardiovascular System - PMC
    The vagus nerve plays an important role in maintaining physiological homeostasis, which includes reflex pathways that regulate cardiac function.
  72. [72]
    Vagus Nerve Stimulation Modulates Complexity of Heart Rate ... - NIH
    This case–control study demonstrated the ability of VNS to increase the complexity of HRV by preferential modulation of the parasympathetic system.
  73. [73]
    Non-invasive vagus nerve stimulation acutely improves ... - PubMed
    May 19, 2017 · tVNS acutely improves spontaneous cardiac baroreflex sensitivity (cBRS) and autonomic modulation, and that these effects are specific to stimulation of ABVN.
  74. [74]
    Transcutaneous auricular vagus nerve stimulation influences gastric ...
    Jun 27, 2021 · ... vagus nerve stimulation. While the vagus nerve's role in gut motility is physiologically established, the effect of taVNS has scarcely been ...
  75. [75]
    The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis
    Feb 7, 2018 · The vagus nerve (VN), the principal component of the parasympathetic nervous system, is a mixed nerve composed of 80% afferent and 20% efferent fibers.
  76. [76]
    Intestinal serotonin acts as a paracrine substance to mediate vagal ...
    We have recently demonstrated that 5-HT released from intestinal enterochromaffin cells activates 5-HT3 receptors on vagal afferent fibres to mediate luminal ...
  77. [77]
    Transcutaneous Auricular Vagus Nerve Stimulation at 20 Hz ...
    Jul 15, 2020 · Transcutaneous Auricular Vagus Nerve Stimulation at 20 Hz Improves Depression-Like Behaviors and Down-Regulates the Hyperactivity of HPA Axis in ...
  78. [78]
    Efficacy and Safety of Transcutaneous Auricular Vagus Nerve ...
    Transcutaneous auricular vagus nerve stimulation (taVNS) is a promising therapy for irritable bowel syndrome (IBS) ... gut motility, gut microbiota ...
  79. [79]
    Evolution of the Vagus Nerve Stimulation (VNS) Therapy System ...
    Implantable Components. Pulse Generator. The VNS Therapy generator is a biologically compatible titanium cased device that uses a lithium carbon monofluoride ...
  80. [80]
    Surgically implanted and non‐invasive vagus nerve stimulation
    These non‐invasive VNS systems improve the safety and tolerability of VNS, making it more accessible and facilitating further investigations across a wider ...
  81. [81]
    Critical Review of Transcutaneous Vagus Nerve Stimulation
    Apr 28, 2020 · The stimulation parameters were set to a frequency of 10 Hz with a pulse width ... pulse widths of the waves, as well as the duty cycle ...
  82. [82]
    Noninvasive Vagus Nerve Stimulation in Postural Tachycardia ...
    Nov 22, 2023 · Active tVNS was performed using a tVNS device (Parasym device; Parasym Health) with an ear clip attached to the tragus (Figure 1), which is ...
  83. [83]
    How I do it: surgical techniques for vagus nerve stimulation in ...
    Feb 20, 2025 · We present a step-by-step description of the VNS implantation technique, incorporating tips and tricks from an epilepsy surgeon with over 20 years of ...
  84. [84]
    Management and outcome of vagus nerve stimulator implantation
    The median duration of surgery was 100 ± 17.1 min (70–165 min) for primary VNS implantation, 45 ± 12.1 min (30–60 min) for generator replacements, and 175.5 ± ...
  85. [85]
    Complications and safety of vagus nerve stimulation - PubMed
    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 ...
  86. [86]
    Vagus Nerve Stimulation Therapy: Indications, Programing, and ...
    Duty cycles can be adjusted up to 49%. However, continuous high frequency (≥ 50 Hz) stimulation could cause nerve injury, and should be avoided (Tables 1, 2).
  87. [87]
    Saudi Arabian Consensus Statement on Vagus Nerve Stimulation ...
    Dec 26, 2020 · Immediate postoperative care. Postoperative wound assessment and potential removal of dressing should be performed before the device activation.
  88. [88]
    Surgical complications of vagus nerve stimulation surgery - PubMed
    Dec 14, 2023 · Results: This study included a total of 606 VNS surgical procedures, leading to 67 complications of which 3 permanent complications.Missing: INSPIRE | Show results with:INSPIRE
  89. [89]
    Delayed vagal nerve compressive neuropathy following placement ...
    Apr 25, 2024 · Postoperative vocal fold paralysis occurs in around 0.26–2.7% of patients implanted with a VNS device. It may be due to excessive intraoperative ...Missing: paresis | Show results with:paresis
  90. [90]
    Intraoperative and perioperative complications with a vagus nerve ...
    A peritracheal hematoma may develop following VNS device placement, necessitating emergent management. Other respiratory complications may include vocal cord ...
  91. [91]
    Complications of vagal nerve stimulation for drug-resistant epilepsy
    Surgical complications were: superficial infection in 3.5%, deep infection needing explantation in 3.5%, vocal cord palsy in 5.6%, which persisted in at least ...
  92. [92]
    Vagal Nerve Stimulation: Indications for Revision in Adult Refractory ...
    Infection, lead fracture, and dual- to single-pin lead battery changes occurred at an incidence of 1.9%, requiring either implant removal or revision. Other ...Missing: migration | Show results with:migration
  93. [93]
    Outcomes following surgical management of vagus nerve stimulator ...
    Dec 18, 2020 · Surgical site infection (SSI) is a rare but significant complication after vagus nerve stimulator (VNS) placement. Treatment options range from ...
  94. [94]
    [PDF] Eliminating Unnecessary Hospital Stays and Prolonged Antibiotics
    A single dose of vancomycin prior to VNS surgery is effective prophylaxis of surgical site infections. Our infection rate of 2% for initial implantation and ...Missing: stimulation | Show results with:stimulation
  95. [95]
    Surgical complications of vagus nerve stimulation surgery
    Complications of VNS surgery include surgical site infection and unilateral vocal cord paresis. Complication rates vary across studies.
  96. [96]
    Revision surgeries following vagus nerve stimulator implantation
    In 1234 patients, average incidence of revision surgeries over 6 years of follow-up were <1%, <3%, 4–10%, and <1% for VNS electrode revision, battery revision/ ...
  97. [97]
    Complications of Implanted Vagus Nerve Stimulation: A Systematic ...
    Mar 12, 2024 · Of note, the most common complications of this technology were voice alteration/hoarseness, paresthesia, pain, increased cough, and dyspnea. The ...
  98. [98]
    Efficacy and tolerability of long-term treatment with vagus nerve ...
    10, 11, 12 Side-effects of VNS treatment are mainly stimulation-related and tend to decrease over time.
  99. [99]
    Bradyarrhythmia secondary to vagus nerve stimulator 7 years after ...
    Jun 30, 2020 · In 0.1% of cases, patients experience bradycardia during initial intraoperative lead testing following VNS device implantation.
  100. [100]
    Aspiration: a potential complication to vagus nerve stimulation
    There needs to be an easy way to turn the device on and off to avoid aspirations, a hazardous and potentially life-threatening complication of VNS.Missing: risk | Show results with:risk
  101. [101]
    Vagus nerve stimulation, side effects, and long-term safety - PubMed
    Common side effects are cough, hoarseness, voice alteration, and paresthesias. These side effects tend to diminish with time. Cognitive side effects often seen ...
  102. [102]
    Single-center long-term results of vagus nerve stimulation ... - PubMed
    May 23, 2023 · The study proved the efficacy and safety of VNS in pediatric patients over an exceptionally long follow-up period.
  103. [103]
    Learnings from 30 years of reported efficacy and safety of vagus ...
    Oct 10, 2020 · VNS efficacy becomes optimal around the sixth month of treatment and a 50–100 % seizure frequency reduction is achieved in approximately 45–65 % of the ...
  104. [104]
    Long-term Expectations of Vagus Nerve Stimulation - PubMed
    VNS battery replacement, revisions, and removals account for almost one-half of all VNS procedures. Our findings suggest important long-term expectations ...
  105. [105]
    Successful removal and reimplant of vagal nerve stimulator device ...
    Our results indicate that the spiral electrodes may be safely removed from the vagus nerve, even after several years. The reversibility of lead implantation may ...
  106. [106]
    Vagal Nerve Stimulation Markedly Improves Long-Term Survival ...
    Therefore, the hepatic effect of vagal stimulation would prevent cardiac remodeling and improve survival of CHF. It is also noted that short-term vagal ...
  107. [107]
    Management of vagus nerve stimulation therapy in the peri ...
    Apr 25, 2023 · Side effects of vagus nerve stimulation therapy include: cough ... Hypotension and bradycardia are common side effects, representing ...
  108. [108]
    Complications and safety of vagus nerve stimulation
    Mar 25, 2016 · The most common complications are postoperative hematoma, infection, and vocal cord palsy.
  109. [109]
    VAGUS-NERVE-STIMULATOR--DEACTIVATION/EXPLANT
    Dec 6, 2003 · Eight (18%) people were identified as having their VNS deactivated or explanted. Records were reviewed from pre-implant through post ...
  110. [110]
    MRI Safety Guidelines for VNS Therapy - LivaNova
    If you have the VNS Therapy® System you may have an MRI safely performed at any MRI center that has a 1.5T or 3T MRI machine and can follow the MRI guidelines.Missing: interactions | Show results with:interactions
  111. [111]
    Safety of a dedicated brain MRI protocol in patients with a vagus ...
    Performing brain MRI scans in patients with an implanted VNS is safe when a modified MRI protocol is followed.Missing: interactions | Show results with:interactions
  112. [112]
  113. [113]
    J.L. Corning and vagal nerve stimulation for seizures in the 1880s
    Feb 12, 2002 · Corning later combined instrumented carotid artery compression with other devices to decrease cerebral blood flow, including transcutaneous ...Missing: Legallois experiments
  114. [114]
    The history of vagotomy - ScienceDirect
    The anatomic characteristics of the vagus nerve were described by Galen in the second century ad, and its physiology was studied by Pavlov almost a century ago.
  115. [115]
  116. [116]
    Peripheral control of hypersynchronous discharge in epilepsy
    Sep 1, 1985 · Peripheral control of hypersynchronous discharge in epilepsy · J. Zabara · Published 1 September 1985 · Medicine · Electroencephalography and ...
  117. [117]
    Inhibition of Experimental Seizures in Canines by Repetitive Vagal ...
    Summary: Repetitive electrical stimulation of the canine cervical vagus nerve interrupts or abolishes motor seizures induced by strychnine and tremors ...
  118. [118]
    Vagus nerve stimulation
    Kiffen Penry, a neurologist, implanted the first VNS device in a 25-year-old man at Wake Forest ... nerve stimulation in patients with refractory epilepsy.
  119. [119]
    Prevention of intractable partial seizures by intermittent vagal ...
    Intermittent stimulation of the vagus nerve in four patients resulted in complete seizure control in two, a 40% reduction of seizure frequency in one.
  120. [120]
    Vagus nerve stimulation in the treatment of epilepsy I
    Stimulation was initiated prior to, or during a seizure episode in dogs. ... Inhibition of experimental seizures in canines by repetitive vagal stimulation.
  121. [121]
    [PDF] Cyberonics, Inc.; Premarket Approval of NeuroCybernetic Prosthesis ...
    SUMMARY: The Food and Drug Administration (FDA) is announcing its approval of the application submitted by Cyberonics, Inc.,. Webster, TX, for premarket ...
  122. [122]
    Vagus Nerve Stimulation Therapy for Epilepsy - IntechOpen
    Jul 16, 2014 · In the 2 randomized, blinded, active-control trials (E03 and E05), patients were randomly assigned to either of 2 treatment groups: HIGH ( ...
  123. [123]
    Vagal nerve stimulator: Evolving trends - PMC - PubMed Central
    History of vagus nerve stimulation (VNS). In 1934, Soma Weiss[2] proposed that compression of the carotid sinus produced a direct cerebral response, causing ...
  124. [124]
    Neuroimmunomodulation of vagus nerve stimulation and the ...
    Jul 6, 2023 · Vagus nerve stimulation (VNS) is a technology that provides ... (HPA) axis-related endocrine functions. The vagal efferent fibers are ...
  125. [125]
    Classification of the External Vagal Nerve Stimulator for Headache
    Dec 27, 2017 · The Food and Drug Administration (FDA or we) is classifying the external vagal nerve stimulator for headache into class II (special controls).Missing: clearance | Show results with:clearance
  126. [126]
    Review of non-invasive vagus nerve stimulation (gammaCore)
    The FDA has cleared gammaCore (non-invasive vagus nerve stimulator [nVNS]) for the treatment of episodic cluster headache (eCH).
  127. [127]
  128. [128]
    Safety and Efficacy of Vagus Nerve Stimulation in Fibromyalgia
    These data suggest that VNS may tune down the pathophysiological processes responsible for central sensitization, thus providing a potential mechanism as to how ...Missing: VAS | Show results with:VAS
  129. [129]
    Vagus Nerve Stimulation Market Size, Trends | Report, 2032
    The global vagus nerve stimulation market size was valued at USD 505.2 million in 2018 and is projected to reach USD 2,294.3 million by 2032, exhibiting a CAGR ...Missing: label | Show results with:label
  130. [130]
    Right‐sided vagus nerve stimulation: Worldwide collection and ... - NIH
    This study highlighted worldwide RS‐VNS outcomes. The key results are enumerated as follows: (1) although off label, we were able to collect more than ...Missing: adoption Asian
  131. [131]
    A closed loop fully automated wireless vagus nerve stimulation system
    Jul 30, 2025 · The primary stimulation parameters of interest are frequency, pulse width (PW), and pulse duty cycle, which are used to reach the neural fulcrum ...
  132. [132]
    Vagus nerve stimulation (VNS): recent advances and future directions
    Oct 4, 2024 · In this trial, 76 patients with resistant epilepsy were randomized to receive either 25 Hz or 1 Hz taVNS. After 20 weeks, responder rates were ...Mechanisms Of Vns · Vns For Epilepsy · Vns For Depression<|control11|><|separator|>
  133. [133]
    Control of spatiotemporal activation of organ-specific fibers in the ...
    May 13, 2025 · We describe a VNS method consisting of intermittent, interferential sinusoidal current stimulation (i 2 CS) through multi-contact epineural cuffs.
  134. [134]
    Feinstein Institutes studies new VNS method | Northwell Health
    May 15, 2025 · Feinstein Institutes research finds new vagus nerve stimulation method targets specific fibers, shows promise for improving therapeutic efficacy.
  135. [135]
    Clinical experience implanting a miniature externally powered ...
    Jun 26, 2025 · A miniature externally powered implantable stimulator (MEPS) that is approximately 50 times smaller than conventional devices, is implanted with a single ...Missing: miniaturization micro- ultrasound charging 2024
  136. [136]
    Best Vagus Nerve Stimulation Device | Top 4 of 2025
    Jun 13, 2025 · We found the four best vagus nerve stimulation devices on the market, tried them ourselves, and studied the science behind their effects.Xen By Neuvana · Pulsetto · Verelief Prime By Hoolest
  137. [137]
    Best Vagus Nerve Stimulation Devices in 2025 - Cybernews
    Oct 23, 2025 · Looking to boost relaxation and focus? Find out the best user-friendly vagus nerve stimulation devices in 2025, how they work and what to ...
  138. [138]
  139. [139]
  140. [140]
    Characterizing the effects of vagus nerve stimulation on symptom ...
    Jul 1, 2025 · The RECOVER trial randomized 493 patients with markedly treatment-resistant major depressive disorder to treatment-as-usual with or without active vagus nerve ...
  141. [141]
    Long-Term Efficacy and Quality-of-Life Changes After Vagus Nerve ...
    Improvements in seizure frequency, duration, and strength were observed in 65% of the patients with drug-resistant epilepsy treated using VNS.
  142. [142]
    Combined minimally invasive vagal cranial nerve and ... - Frontiers
    Aug 25, 2025 · The significant improvements observed in our observational study include an 87% reduction in pain, a 37% decrease in mean blood glucose, a ...
  143. [143]
    Transcutaneous vagus nerve stimulation improves Long COVID ...
    May 1, 2024 · This pilot study provides preliminary evidence supporting the potential of t-VNS as a therapeutic intervention for female Long COVID patients.
  144. [144]
    Non-invasive vagus nerve stimulation and exercise capacity in ...
    Feb 19, 2025 · Non-invasive vagal stimulation improves measures of cardiorespiratory fitness and attenuates inflammation, offering an inexpensive, safe, and ...Non-Invasive Vagus Nerve... · Methods · Transcutaneous Vagus Nerve...