Next Article in Journal
The Evaluation of Oral Health Condition and Oral and Dental Care in Children with Congenital Heart Disease
Previous Article in Journal
Comparative Analysis of Hematological and Immunological Parameters in Patients with Primary Sjögren’s Syndrome and Peripheral Neuropathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Effects of Non-Invasive Neuromodulation of the Vagus Nerve for Management of Tinnitus: A Systematic Review with Meta-Analysis

by
David Fernández-Hernando
1,*,
Cesar Fernández-de-las-Peñas
2,
Ana Machado-Martín
3,
Santiago Angulo-Díaz-Parreño
4,
Francisco J. García-Esteo
1,4 and
Juan A. Mesa-Jiménez
1,4
1
Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
2
Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
3
Servicio de Otorrino-Laringología, Hospital Universitario Quiron Salud Pozuelo, Universidad Europea de Madrid, 28670 Madrid, Spain
4
Facultad de Medicina, Universidad San Pablo CEU, 28660 Madrid, Spain
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(11), 3673; https://doi.org/10.3390/jcm12113673
Submission received: 23 April 2023 / Revised: 16 May 2023 / Accepted: 23 May 2023 / Published: 25 May 2023
(This article belongs to the Section Otolaryngology)

Abstract

:
Background: Tinnitus is the perception of sound in the absence of actual external stimuli. Other associated symptoms include frustration, annoyance, anxiety, depression, stress, cognitive dysfunction, insomnia, or emotional exhaustion. Objective: In this study, we aimed to conduct a systematic review and meta-analysis on the effectiveness of the non-invasive neuromodulation of the vagus nerve in patients with tinnitus. Methods: Six databases were searched from their date of inception to 15 June 2022 to identify clinical trials in which at least one group received any form of non-invasive neuromodulation of the vagus nerve for tinnitus management, with outcomes based on annoyance and related disability. Data on participants, interventions, blinding strategies, assessment outcomes, and results were extracted by two reviewers. Results: The search identified 183 articles with five clinical trials eligible for inclusion in the review and four for the meta-analysis. The methodological quality scores ranged from 6 to 8 (mean: 7.3, SD: 0.8) points. The meta-analysis identified a significant positive effect on THI post-treatment for unilateral auricular stimulation (hg = 0.69, 95% CI 0.06, 1.32) or transcutaneous nerve stimulation (hg = 0.51, 95% CI 0.1, 0.9) compared with a comparative group. No effect on loudness intensity was observed. Conclusion: The results of the meta-analysis suggest that the application of the non-invasive neuromodulation of the vagus nerve has a positive effect post-treatment in terms of related disability in patients with tinnitus, although its clinical relevance is low. No firm conclusions about the effect of the non-invasive neuromodulation of the vagus nerve on tinnitus are available based on the current literature.

Graphical Abstract

1. Introduction

Auricular transcutaneous vagus nerve stimulation (at-VNS) is a modality of non-invasive nerve stimulation (n-VNS), commonly used in clinical practice to treat tinnitus (acute or chronic), cluster headaches, migraines, depression, epilepsy, and other disorders, as well as atrial fibrillation, prosocial behavior, associative memory, schizophrenia, or pain [1,2,3,4]. n-VNS is a safe, reliable treatment with no adverse effects in response to medication intake [4,5]. At-VNS is a non-invasive and non-expensive therapy that involves stimulating the auricular branch of the vagus nerve (ABVN) at the outer parts of the ear, conferring autonomic benefits [6,7,8]. One of the main differences between VNS and at-VNS is that, in at-VNS, patients do not need general anesthesia for implantation, making this treatment safer than VNS [3,4]. The former method is also more expensive and riskier than at-VNS, with costs ranging from USD 30,000 to USD 50,000 [5,9]. Magnetic resonance imaging (MRI) has recently highlighted an anatomical pathway for at-VNS, for example, through the neck and auricula, which connect the nervous system and the external parts of the body [7,8,10]. The physiological effects of at-VNS on the brain have yet to be fully elucidated, and studies are not homogeneous in their results due to the high risk of bias and unclear parameters, stimulation intensity, pulse width, waveform, or frequency.
Tinnitus is the perception of sound in the absence of actual external sounds. It has an annual prevalence of 1%, being experienced by 14% of the population. Tinnitus is not a severe otological symptom in most cases, and only 2% of cases are severe [11,12,13,14]. Its prevalence does not differ by sex but increases with age, affecting 10% of young adults, 14% of middle-aged people, and 24% of adults as a whole [11,14]. Symptoms include annoyance, anxiety, depression, cognitive dysfunction, insomnia, and emotional exhaustion. Patients with tinnitus experience hearing loss, with ipsilateral tinnitus on one side being the most reported pathology [11,12,15]. A possible explanation for tinnitus is that the damage to the inner ear involving hair cells causes hearing loss, and the input enters the cochlea, with the latter sending incorrect information to the superior level on the brain cortex—more specifically, the auditory cortex. Physical tests such as audiograms may be normal in these patients [10,11,12,15,16].
Recently, a systematic review on the treatment of tinnitus with n-VNS including some studies with invasive treatment was published, differing completely from this systematic review, which only includes non-invasive neuromodulation; however, no meta-analysis was conducted, and it included only two randomized controlled trials (RCTs), five cohorts studies, and two case series [17]. Meanwhile, this systematic review and meta-analysis included randomized controlled trials (RCTs) as well as controlled trials that related the quality of studies and the effectivity of n-VNS as a treatment for tinnitus with a novel technique, at-VNS, distinguishing them completely from all the RCTs included in both systematic reviews. Furthermore, a recent study provides evidence applied in rats that VNS is more useful and safer in combination with drugs to prevent adverse events and that VNS therapy mediates plasticity and enhances recovery as part of neuro-rehabilitation [18].

2. Materials and Methods

A systematic review and meta-analysis following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines was conducted [19]. This review was prospectively registered in the PROSPERO database (registration number: CRD42021265126).

2.1. Search Strategy

Electronic literature searches were conducted on the CINALH, MEDLINE, SCOPUS, PUB MED, PEDro, and EMBASE databases from their inception to 15 June 2022. When the searched databases allowed limits, searches were restricted to randomized clinical trials. We also screened the reference lists of the papers identified in the searches. The search strategy included the following MeSH terms: “Vagus Nerve”, “Auricular Vagus Nerve Stimulation”, “Transcutaneous Vagus Nerve Stimulation”, and “Tinnitus”. These terms were combined with “AND” or “OR” operators, as follows:
((Non-Invasive neuromodulation) OR (auricular Transcutaneous Vagus Nerve Stimulation) OR (Transcutaneous Vagus Nerve Stimulation)) AND ((Tinnitus) OR (Chronic Tinnitus)).

2.2. PICO Principle

This review/meta-analysis was conducted to answer the following clinical question: Is it effective to use at-VNS to treat tinnitus?
Population, Intervention, Comparison, and Outcome (PICO) questions.
Population: Adults older than 18 years of age with chronic tinnitus or severe tinnitus.
Intervention: Application of auricular transcutaneous vagus nerve stimulation technique.
Comparator: Acceptable comparators were any type of placebo (e.g., turning off device), sham, or no intervention.
Outcomes: The primary outcome measure was tinnitus annoyance (assessed by a visual analogue scale, VAS, or numerical scale), related disability (assessed by the Tinnitus Handicap index, THI), or hearing loss (audiometry, pure tone average).

2.3. Study Selection

Inclusion criteria: (i) Population: studies with patients of any age or sex with a chronic or severe tinnitus diagnosis made by a health professional. (ii) Application of auricular transcutaneous vagus nerve stimulation technique. (iii) Comparison: Acceptable comparators were any type of placebo (e.g., turning off device), sham, or no intervention. (iv) Outcome: The primary outcome measure was tinnitus annoyance (assessed by a visual analogue scale, VAS, or numerical scale), related disability (assessed by the Tinnitus Handicap Index, THI), or hearing loss (audiometry, pure tone average). (v) Studies: This review included randomized controlled, clinical trials, controlled trials, and pilot clinical trials since these designs were suitable to answer our question.
Exclusion criteria: (i) Population: studies whose patients had no evidence of any tinnitus diagnosis. (ii) Intervention/exposure/factor: studies that used surgical interventions of VNS. (iii) Comparison: studies with no comparison group. (iv) Studies: systematic reviews, meta-analyses, reviews, case reports, letters to the editor, conference articles, book chapters, protocol registries, grey literature, cadaver studies, and animal studies.

2.4. Data Extraction and Quality Assessment

Data extraction was performed by two authors, and the data were compiled into a standardized data extraction form in an excel spreadsheet. Data included simple size, diagnosis, inclusion/exclusion criteria, duration of symptoms, intervention (location, technique, and duration), main outcomes, follow-ups, and adverse events. In case of a discrepancy between authors, an agreement had to be achieved; a third author was in charge of reaching a consensus.
Methodological quality was evaluated using the PEDro (Physiotherapy Evidence Database) scale and the ROB-2 Cochrane tool; this process was carried out independently by two authors. The RoB-2 tool includes the following items: selection bias (randomization sequence generation, allocation concealment), performance bias (blinding participants, blinding therapists), detection bias (blinding outcome assessor), attrition bias (incomplete outcome data), reporting bias (source of funding bias/selecting outcome reporting), and other bias (sample size) [20]. Each item was classified as low risk, high risk, or unclear, according to the Cochrane Collaboration tool [20]. In all cases, an answer ‘Yes’ indicated a low risk of bias, and an answer ‘No’ indicated a high risk of bias. If insufficient details were reported regarding what occurred during the trial or the entry was not relevant to the study (particularly for assessing blinding and incomplete outcome data, when the outcome being assessed by the entry had not been measured in the study) the answer was ‘unclear’ risk of bias [20,21].
The PEDro scale is based on 11 criteria, of which 10 contribute to the score, representing methodological quality. The PEDro scale has been shown to have fair to good interrater reliability (ICC 0.55, 95% CI 0.41–0.72) [22]. The scale assessed the following items: random allocation; concealed allocation; between-groups similarity at baseline; participant blinding; therapist blinding; assessor blinding; dropout; intention-to-treat statistical analysis; between-group statistical comparison; point measures; and variability data. A PEDro score ≥ 5/10 points determines a high-quality trial.

2.5. Level of Evidence

We used the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach [23]. According to the GRADE approach, the level of evidence can be classified as high quality (the authors are very confident that the intervention effect is close to the estimated effect), moderate quality (the authors are somewhat confident that the intervention effect is probably close to the estimated effect, but there is a possibility that it is different), and low quality (the intervention effect may be markedly different from the estimated effect) [23]. GRADE assesses quality according to four main domains: risk of bias, inconsistency, indirectness, and imprecision. The overall classification of the evidence is assessed as either “high”, “moderate”, “low”, or “very low”. For risk of bias, we established that a “very low” quality rating required a study to receive a ≥2 high risk of bias rating or 1 specific high risk rating in the “other category”, plus a >2 unclear rating or 3 unclear rating in dimensions other than the “other category” using the Cochrane risk of bias tool. A “low” quality rating required that a study receive at least 1 high risk rating; a “moderate” quality rating required that the study received 2 unclear risk of bias ratings in a category related to randomization and/or blinding; and a “high” quality rating required that a study receive 1 or fewer unclear risks of bias.

2.6. Data Synthesis and Analysis

We extracted the sample size, means, and standard deviations for each variable. When the trial reported standard error or non-parametric values (median or interquartile range), they were converted to standard deviations and parametric values as needed. If necessary, the mean scores and standard deviations were estimated from graphs.
The between-groups mean differences of the trials were converted to effect sizes using a random-effects model. An effect size of 0.8 or greater was considered large, between 0.5 and 0.8 was considered moderate, and between 0.2 and 0.5 was considered small. p-values < 0.05 were considered statistically significant. Due to the small number of studies identified in the review, the meta-analytic comparison was based on the mean difference change (hg) in the tinnitus variable (THI, VAS loudness, or VAS uncomfortable) at post-treatment and in the short term (four-weeks) after at-VNS compared with the control or comparative group.
For inconsistency, we used I2 heterogeneity between studies, estimated using Cochrane. The Cochrane group establishes the following interpretation of the I2 statistic: 0–40% may represent irrelevant/unimportant heterogeneity, 30–60% suggests moderate heterogeneity, 50–90% represents substantial heterogeneity, and 75–100% represents considerable heterogeneity [24]. As described by the Cochrane group, the data of heterogeneity overlap. The importance of the observed value of I2 is based on the magnitude and the direction of effects and the strength of evidence of heterogeneity (p value). In grading the imprecision of continuous outcomes, the effect size magnitude cutoffs proposed by Cohen were used as benchmarks for assessing the magnitude of the confidence intervals of observed effects. For example, 95% confidence intervals (CI) of a standardized mean difference (SMD) extending between >0.2 and <0.5 points in either direction indicate a “serious” quality downgrade; 95% CI of an SMD extending >0.5 point in either direction indicate a “very serious” quality downgrade [25].
Publication bias was assessed graphically using funnel plots and the Egger regression test [23]. Statistical assessment was two-tailed and was considered statistically significant at p < 0.05 [25,26,27]. The sensitivity analysis was performed using MIX 2.0 Pro.

3. Results

3.1. Study Selection

The data search yielded a total of 183 articles, including duplicates. Thirty-eight articles were included for abstract/full text review, five were included in the systematic review [28,29,30,31,32], and four were included in the meta-analysis [29,30,31], while three were excluded based on the inclusion and exclusion criteria [33,34,35]. Figure 1 shows the flow diagram of the data search and the studies that met the inclusion/exclusion criteria.

3.2. Study Characteristics

The total sample size consisted of 234 participants (age: 47, 93, SD: 11, 49 years, 61.96% men). The duration of tinnitus-associated symptoms ranged from 3 months to 2 years (mean: 51.3, SD: 58 weeks). The article types included three RCTs [29,30,31], one clinical controlled trial [32], and one clinical trial [28], and there were three excluded studies, which did not report the tinnitus duration or hearing loss threshold or the article type [33,34,35] (Table 1). All the studies included in this systematic review at baseline characteristics measured tinnitus duration and audiological assessment of hearing loss threshold (pure tone audiometry, tympanometry, speech discrimination score).

3.3. Outcomes and Follow-Ups

We extracted the following outcomes from the THI [29,30,31,32] and VAS: Loudness, annoyance, or discomfort [29,30,31] were the most commonly assessed, while others included pure tone average (PTA), speech discrimination [28,29,31,32], depression anxiety stress scales (DASS) [29], audiometry [28,29,30,31,32], MRI for cortical activation [32], heart rate, vagal somatosensory evoked potential test, minimum masking level (MML), tinnitus loudness matching (TLM), audiograms, and pure tone thresholds [29].
Five studies evaluated outcomes immediately after the last treatment session [28,29,31,32,35], whereas two trials evaluated the effects in the short term (4 weeks after) [30,31].

3.4. Interventions/Control Groups

For the intervention group, it is important to underline the types of modalities including at-VNS. Some studies used a turn-on Transcutaneous Electrical Nerve Stimulation (TENS) in the auricular branch of the vagus nerve [29,30,31,32]. Another type of intervention used monomodal versus bi-modal somatosensory stimulation [28].
The parameters of electrical stimulation were inconsistent in frequency, interval time, or treatment time among trials. While there were four trials with similar parameters in terms of current frequency (i.e., 20–30 Hz) [29], the pulse width, treatment, and electrodes placement were consistently different. If the area of application was similar, the position did not remain the same, even if it depended on the type of intervention or study protocol. The most common areas for electrode placement were the inner tragus, cymba conchae, C2 dermatome, triangular fossa, inner ear, meatus, and tragus [29,30,31,32].
The control group in the studies exhibited the following factors: turn-off devices for sham at-VNS [29,32]; anatomical regions other than the ear lobe combined with manual acupuncture [30,31]; another, different intervention [28].

3.5. Methodological Quality and Risk of Bias

The methodological quality scores ranged from 6 to 8 (mean: 7.3, SD: 0.8) out of a maximum of 10 points (Table 2). Only three studies were of high methodological quality (≥5 points) [29,30,31], as shown in Figure 2. The ROB-2 Cochrane tool demonstrated that all studies had high risk of bias in items, except for two RCTs that had low risk [30,31]. The others all had high risk [28,29,32]. The overall results show us 40% of low risk and 60% of high risk of bias in the assessment. For each domain the assessment was D1; 60% for low risk and 40% for high risk of bias, D2; 20% for low and high risk, respectively, and 60% for moderate risk of bias, D3; 80% for low risk and 20% for high risk of bias, D4; 60% for moderate and 40% for high risk of bias, and D5; 20% for low risk and 80% for moderate risk of bias.

3.6. Unilateral Auricular Stimulation vs. Control

The meta-analysis revealed a significant effect on THI (hg = 0.69, 95% CI 0.06 to 1.32, Z = 2.15, p = 0.04, I2 = 56%, n = 3, Figure 3A) but not on VAS loudness (hg −0.01, 95% CI −0.47 to 0.45, Z = −0.04, p = 0.97, I2 = 0%, n = 2, Figure 3B) or VAS uncomfortable (hg = 0.31, 95% CI −0.34 to 0.96; Z = 0.95, p = 0.34, I2 = 32%, n = 2, Figure 3C) post-treatment.
No significant effect on THI (hg = 0.28, 95% CI −0.25 to 0.81; Z = 1,03, p = 0.3, I2 = 0%, n = 2), VAS loudness (hg = −0.02, 95% CI −0.48 to 0.44; Z = −0.1, p = 0.92, I2 = 0%, n = 2), or VAS uncomfortable (hg = 0.31, 95% CI −0.16 to 0.77; Z = 1.29; p = 0.2; I2 = 0%, n = 2) at short-term was found either.

3.7. Unilateral Auricular Stimulation vs. Bilateral Stimulation

The meta-analysis revealed a non-significant effect on THI (hg = 0.21, 95% CI −0.13 to 0.55; Z = 1.21, p = 0.23, I2 = 0%, n = 4), VAS loudness (hg = 0.19, 95% CI −0.34 to 0.71, Z = 0.7, p = 0.49, I2 = 0%, n = 2), and VAS uncomfortable (hg = 0.15, 95% CI −0.38 to 0.67, Z = 0.55, p = 0.58, I2 = 0%, n = 2) post-treatment. There was no significant effect on THI (hg = 0.3, 95% CI −0.22 to 0.83; Z = 1.13, p = 0.26, I2 = 0%, n = 2), VAS loudness (hg = 0.13, 95% CI −0.39 to 0.65, Z = 0.48, p = 0.63, I2 = 0%n = 2), or VAS uncomfortable (hg = 0.16, 95% CI −0.36 to 0.67, Z = 0.59, p = 0.55, I2 = 0%, n = 2) in the short term.

3.8. TENS vs. Control

The meta-analysis revealed a significant effect on THI (hg = 0.51, 95% CI 0.1 to 0.9, Z = 2.45, p = 0.01, I2 = 0%, n = 3), but not on VAS loudness (hg = 0.19, 95% CI −0.34 to 0.71, Z = 0.7, p = 0.49, I2 = 0%, n = 2) or on VAS uncomfortable (hg = 0.07, 95% CI −0.45 to 0.59, Z = 0.27, p = 0.79, I2 = 0%, n = 2) post-treatment. No significant effects on THI (hg = 0.3, 95% CI −0.22 to 0.83, Z = 1.14, p = 0.26, I2 = 0%), VAS loudness (hg = 0.13, 95% CI −0.39 to 0.65, Z = 0.48, p = 0.63, I2 = 0%, n = 2), or VAS uncomfortable (hg = 0.16, 95% CI −0.36 to 0.68, Z = 0.6, p = 0.55, I2 = 0%, n = 2) were observed in the short term. Quality of Evidence (GRADE).
The low quality of evidence (heterogeneity) must be taken carefully because of the small number of studies available and the low quality of some of them in terms of the effectiveness of non-invasive neuromodulation for tinnitus in the short and long term (Table 3).

4. Discussion

The objective of this meta-analysis was to determine the effects of non-invasive neuromodulation of the vagus nerve for the management of tinnitus-associated symptoms. The results suggest that the application of non-invasive neuromodulation of the vagus nerve has a positive post-treatment effect on related disability in patients with tinnitus, although its clinical relevance is small.
Certain points are in need of further consideration, as a recent systematic review and an n-NVS-related review published in recent years [17] both revealed slightly different findings. The first difference was in the eligibility criteria, since we only included clinical trials, whereas Stegeman et al. [17] included case reports, case series, cohorts studies, and only two RCTs. The inclusion of just clinical trials allowed us to pool data and conduct the first meta-analysis. Secondly, the assessment of methodological quality or risk of bias revealed a high risk of bias and several concerns regarding the quality of trials [28,29,32,33,34,35]. These studies also have a control group which can be used to compare the interventions with turn-off devices or different interventions or to compare the same intervention involving different anatomical spaces [28,29,30,31,32,33,34,35]. In the last publication, there were no comparisons, indicating that there is a high risk of bias regarding conclusions [17], with one exception being the RCT crossover study that was excluded from our systematic review because it had an invasive intervention [17,36]. Additionally, another important difference was that the last publication divided into VNS with paired sound and VNS without paired sound including a unique study where at-VNS was used, but that was a cohort prospective study [17].
Recently, VNS in rats was proven to be a remarkable neuro-rehabilitation therapy because it influences network connectivity and motor control. It releases pro-plasticity factors such as norepinephrine, acetylcholine, serotonin, brain-derived neurotrophic factor, and fibroblast growth factor [37]. The majority of them were identified in spinal cord injuries or brain injuries in rat models. However, these studies demonstrate the pathways and how neuroplasticity works. VNS therapy can stimulate muscles, nerves, and the brain; thus, it could help to treat pathologies such as spinal cord injuries because it strengthens the connectivity between them and assists with recovery. Furthermore, VNS therapy takes more evidence in the activation of cholinergic and noradrenergic modulatory pathways [38,39,40]. Furthermore, these new advances could have application in humans as a new treatment for spinal cord injuries or any neurological disorders such as tinnitus.
In the last year, findings have emerged which support the new advances in neuromodulation to treat neurological disorders. These techniques with an implant demonstrate the relationship between peripheral nerves and the central nervous system. It is important to underline that they are their safety and reliability as a neuromodulation treatment has not yet been proven. They are more accurate than other treatments in terms of the specific parameters, with greater efficacy [39]. There are also non-invasive devices which represent a new clinical treatment option, as demonstrated in recent clinical trials; these devices induce tactile stimulation of the Vagus nerve using a VNS implantation. This technique has provided evidence of enhanced peripheral stimulation, suggesting that neuroplasticity can be used to recover motor functions after brain damage [41].
This study also identified heterogeneity in the parameters of the interventions in terms of frequency, intensity, and wave pulse, which made it difficult to determine which one was effective. Simultaneously, the number of sessions, position of electrodes, and duration of treatment were also different, and a consensus is therefore necessary. Further, randomized clinical trials must be contemporary to guarantee their methodological quality [42,43,44,45]. For example, the latest publications that explore using acupuncture alone to treat tinnitus have the same results after the operation and three-weeks postoperatively, demonstrating that follow-up for this pathology has to be applied effectively to determine the presence of a positive or long-term effect [46].
Few trials have investigated the pathways of tinnitus, migraine, and epilepsy and the underlying mechanisms of how non-invasive neuromodulation of the vagus nerve could work [3,13,47]. This peripheral stimulation of the nervous system tries to change the central nervous system without adverse events, thus increasing the chances of successful treatment. Trying to understand the main pathways and mechanisms of action with which to treat these types of pathologies is a topic for further research. Additionally, not all studies followed the same protocol with the same parameters of the point of application of at-NVS, or ABVNS, based on this principle, which may be a different path of treatment.
The follow-ups for measurements are inconsistent in all studies compared with the last publication [17]. Moreover, the results are the same immediately post-treatment [28,29,30,31,32,33,34,35] and four weeks later [30,31]. The lack of long-term follow-up periods makes it harder to draw a conclusion.
In the meta-analysis, we have to make a strong critique regarding the low quality and the small number of studies because there were only three RCTs of high methodological quality (PEDro score > 5 points). The data provided were not the most suitable for comparison due to their heterogeneity [29,30,31], and most studies presented some concerns in relation to ROB-2 in the final assessment [32]. Even if the results are slightly positive for interventions, it is the same before intervention, and low THI increases the risk of bias and affects quality. For those reasons, we cannot draw any firm conclusion about the true effectiveness of at-VNS for the management of tinnitus. Moreover, it is important to underline the relation between hearing loss threshold and audiological examination at baseline and the results of THI since it is the main symptom of tinnitus and has shown the most improvement after treatment using at-VNS compared with control groups. Additionally, because patients with tinnitus have this chronic otological symptom related to hearing loss, they are classified as having severe or chronic tinnitus based on the duration of symptoms and age extracted from all studies included in this systematic review.Furthermore, slight improvement in hearing loss could have positive effects on other symptoms such as anxiety and the quality of life or life style of many suffering patients. Because of the previous critiques the improvement of the methodological study design is mandatory, and should incorporate the use of similar follow-ups and parameters to be able to determinate if at-VNS can improve this symptom based on the hearing loss threshold examination and consequently improve the quality of life of patients who suffer from chronic or severe tinnitus.
One of the main limitations of this meta-analysis is the small number of studies available and their poor methodological quality [29,30,31]. Most outcomes were no different for the intervention at any follow-up period. There were differences only in THI post-treatment, with some positive results in the TENS and unilateral auricular stimulation groups. Consequently, it is mandatory to provide RCTs with a better methodology and low risk of bias to provide strong evidence of the possible effectiveness of at-NVS in patients with tinnitus. Another limitation was the difference between parameters, the position of the electrodes, and protocols of the studies of at-VNS [30,31,32,33] and at-VNS [29,30,31,32,33,35] or bi-phasic stimulation [28], which made it difficult to verify which one is more effective for treating tinnitus.

5. Conclusions

The current meta-analysis on at-VNS found that non-invasive neuromodulation of the vagus nerve could have a slightly positive post-treatment effect on THI outcomes in patients with tinnitus, but its clinical relevance is small because of the number of studies available for the meta-analysis. The number of trials was small, and the studies presented high risk of bias. No conclusion about the effect of non-invasive neuromodulation of the vagus nerve on tinnitus is available based on the current literature. Of these studies, there were none of sufficient quality to asseverate the findings, and high-quality studies must be conducted in the future.

Author Contributions

J.A.M.-J. contributed to the conception and design of this study. S.A.-D.-P. contributed to the analyses of the data. All authors contributed to the interpretation of the data. Article drafts were written by D.F.-H. and J.A.M.-J. and critically revised by all authors. The final version of the article was approved by all authors. J.A.M.-J. takes responsibility for the integrity of the work as a whole. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Prospero number; CRD42021265126.

Informed Consent Statement

Not applicable.

Data Availability Statement

We analyzed our meta-analysis using an excel spreadsheet with the program Mix 2.0 Pro. We extracted the sample size, means, and standard deviations for each variable. When the trial reported standard error or non-parametric values (median or interquartile range), they were converted to standard deviations and parametric values as needed. If necessary, the mean scores and standard deviations were estimated from graphs. The between-groups mean differences of the trials were converted to effect sizes using a random-effects model. An effect size of 0.8 or greater was considered large, between 0.5 and 0.8 as moderate, and between 0.2 and 0.5 as small. p-values < 0.05 were considered statistically significant.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Li, T.-T.; Wang, Z.-J.; Yang, S.-B.; Zhu, J.-H.; Zhang, S.-Z.; Cai, S.-J.; Ma, W.-H.; Zhang, D.-Q.; Mei, Z.-G. Transcutaneous electrical stimulation at auricular acupoints innervated by auricular branch of vagus nerve pairing tone for tinnitus: Study protocol for a randomized controlled clinical trial. Trials 2015, 16, 1–9. [Google Scholar] [CrossRef] [PubMed]
  2. Amiri, S.; Behnezhad, S.; Azad, E. Migraine headache and depression in adults: A systematic Review and Meta-analysis. Neuropsychiatrie 2019, 33, 131–140. [Google Scholar] [CrossRef] [PubMed]
  3. Yap, J.Y.Y.; Keatch, C.; Lambert, E.; Woods, W.; Stoddart, P.R.; Kameneva, T. Critical Review of Transcutaneous Vagus Nerve Stimulation: Challenges for Translation to Clinical Practice. Front. Neurosci. 2020, 14, 284. [Google Scholar] [CrossRef]
  4. Badran, B.W.; Dowdle, L.T.; Mithoefer, O.J.; LaBate, N.T.; Coatsworth, J.; Brown, J.C.; DeVries, W.H.; Austelle, C.W.; McTeague, L.M.; George, M.S. Neurophysiologic effects of transcutaneous auricular vagus nerve stimulation (taVNS) via electrical stimulation of the tragus: A concurrent taVNS/fMRI study and review. Brain Stimul. 2018, 11, 492–500. [Google Scholar] [CrossRef] [PubMed]
  5. Benemei, S.; Labastida-Ramírez, A.; Abramova, E.; Brunelli, N.; Caronna, E.; Diana, P.; Gapeshin, R.; Hofacker, M.D.; Maestrini, I.; Pías, E.M.; et al. Persistent post-traumatic headache: A migrainous loop or not? The preclinical evidence. J. Headache Pain 2020, 21, 90. [Google Scholar] [CrossRef] [PubMed]
  6. Choi, W.S.; Kim, S.J.; Chang, D.S.; Lee, H.Y.; Suk, W.C. Characteristics of Stimulus Intensity in Transcutaneous Vagus Nerve Stimulation for Chronic Tinnitus. J. Int. Adv. Otol. 2018, 14, 267–272. [Google Scholar] [CrossRef]
  7. Kaniusas, E.; Kampusch, S.; Tittgemeyer, M.; Panetsos, F.; Gines, R.F.; Papa, M.; Kiss, A.; Podesser, B.; Cassara, A.M.; Tanghe, E.; et al. Current Directions in the Auricular Vagus Nerve Stimulation I–A Physiological Perspective. Front. Neurosci. 2019, 13, 1–23. [Google Scholar] [CrossRef]
  8. Zhang, Y.; Liu, J.; Li, H.; Yan, Z.; Liu, X.; Cao, J.; Park, J.; Wilson, G.; Liu, B.; Kong, J. Transcutaneous auricular vagus nerve stimulation at 1 Hz modulates locus coeruleus activity and resting state functional connectivity in patients with migraine: An fMRI study. NeuroImage Clin. 2019, 24, 101971. [Google Scholar] [CrossRef]
  9. Richards. 乳鼠心肌提取 HHS Public Access. Physiol. Behav. 2018, 176, 139–148. [Google Scholar]
  10. Sclocco, R.; Garcia, R.G.; Kettner, N.W.; Isenburg, K.; Fisher, H.P.; Hubbard, C.S.; Ay, I.; Polimeni, J.R.; Goldstein, J.; Makris, N.; et al. The influence of respiration on brainstem and cardiovagal response to auricular vagus nerve stimulation: A multimodal ultrahigh-field (7T) fMRI study. Brain Stimul. 2019, 12, 911–921. [Google Scholar] [CrossRef]
  11. Kim, H.-J.; Lee, H.-J.; An, S.-Y.; Sim, S.; Park, B.; Kim, S.W.; Lee, J.S.; Hong, S.K.; Choi, H.G. Analysis of the Prevalence and Associated Risk Factors of Tinnitus in Adults. PLoS ONE 2015, 10, e0127578. [Google Scholar] [CrossRef]
  12. Rhee, J.; Lee, D.; Suh, M.-W.; Lee, J.H.; Hong, Y.-C.; Oh, S.H.; Park, M.K. Prevalence, associated factors, and comorbidities of tinnitus in adolescents. PLoS ONE 2020, 15, e0236723. [Google Scholar] [CrossRef] [PubMed]
  13. McCormack, A.; Edmondson-Jones, M.; Somerset, S.; Hall, D. A systematic review of the reporting of tinnitus prevalence and severity. Hear. Res. 2016, 337, 70–79. [Google Scholar] [CrossRef]
  14. Jarach, C.M.; Lugo, A.; Scala, M.; Brandt, P.A.V.D.; Cederroth, C.R.; Odone, A.; Garavello, W.; Schlee, W.; Langguth, B.; Gallus, S. Global Prevalence and Incidence of Tinnitus: A Systematic Review and Meta-analysis. JAMA Neurol. 2022, 79, 888–900. [Google Scholar] [CrossRef]
  15. Bhatt, J.M.; Bhattacharyya, N.; Lin, H.W. Relationships between tinnitus and the prevalence of anxiety and depression. Laryngoscope 2017, 127, 466–469. [Google Scholar] [CrossRef]
  16. Lehtimäki, J.; Hyvärinen, P.; Ylikoski, M.; Bergholm, M.; Mäkelä, J.P.; Aarnisalo, A.; Pirvola, U.; Mäkitie, A.; Ylikoski, J. Transcutaneous vagus nerve stimulation in tinnitus: A pilot study. Acta Oto-Laryngologica 2013, 133, 378–382. [Google Scholar] [CrossRef] [PubMed]
  17. Stegeman, I.; Velde, H.M.; Robe, P.A.J.T.; Stokroos, R.J.; Smit, A.L. Tinnitus treatment by vagus nerve stimulation: A systematic review. PLoS ONE 2021, 16, e0247221. [Google Scholar] [CrossRef] [PubMed]
  18. Morrison, R.A.; Abe, S.T.; Danaphongse, T.; Ezhil, V.; Somaney, A.; Adcock, K.S.; Rennaker, R.L.; Kilgard, M.P.; Hays, S.A. Common Cholinergic, Noradrenergic, and Serotonergic Drugs Do Not Block VNS-Mediated Plasticity. Front. Neurosci. 2022, 16, 849291. [Google Scholar] [CrossRef]
  19. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  20. Higgins, J.P.T.; Altman, D.G.; Gøtzsche, P.C.; Jüni, P.; Moher, D.; Oxman, A.D. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 2011, 343, d5928. [Google Scholar] [CrossRef]
  21. Sterne, J.A.C.; Savović, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef]
  22. Maher, C.G.; Sherrington, C.; Herbert, R.D.; Moseley, A.M.; Elkins, M. Reliability of the PEDro Scale for Rating Quality of Randomized Controlled Trials. Phys. Ther. 2003, 83, 713–721. [Google Scholar] [CrossRef]
  23. Schünemann, H.J.; Oxman, A.D.; Brozek, J.; Glasziou, P.; Bossuyt, P.; Chang, S.; Muti, P.; Jaeschke, R.; Guyatt, G.H. GRADE: Assessing the quality of evidence for diagnostic recommendations. BMJ Evidence-Based Med. 2008, 13, 162–163. [Google Scholar] [CrossRef]
  24. Deeks, J.J. Chapter 9: Analyzing data and undertaking metanalyses. In Cochrane Handbook for Systematic Reviews of Interventions, version 520 (updated June 2017); Higgins, J.P.T., Churchill, R., Chandler, J., Cumpston, M.S., Eds.; Cochrane: Singapore, 2017; Available online: www.training.cochrane.org/handbook (accessed on 15 October 2022).
  25. DerSimonian, R.; Laird, N. Meta-analysis in clinical trials. Control. Clin. Trials 1986, 7, 177–188. [Google Scholar] [CrossRef]
  26. Ades, A.E.; Lu, G.; Higgins, J.P.T. The Interpretation of Random-Effects Meta-Analysis in Decision Models. Med. Decis. Mak. 2005, 25, 646–654. [Google Scholar] [CrossRef]
  27. Higgins, J.P.T.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef] [PubMed]
  28. Hamilton, C.; D’arcy, S.; Pearlmutter, B.A.; Crispino, G.; Lalor, E.C.; Conlon, B.J. An Investigation of Feasibility and Safety of Bi-Modal Stimulation for the Treatment of Tinnitus: An Open-Label Pilot Study. Neuromodulation 2016, 19, 832–837. [Google Scholar] [CrossRef]
  29. Tutar, B.; Atar, S.; Berkiten, G.; Üstün, O.; Kumral, T.L.; Uyar, Y. The effect of transcutaneous electrical nerve stimulation (TENS) on chronic subjective tinnitus. Am. J. Otolaryngol. 2019, 41, 102326. [Google Scholar] [CrossRef] [PubMed]
  30. Kim, B.H.; Moon, Y.K.; Kim, M.H.; Nam, H.J. Comparing the effects of manual acupuncture, electroacupuncture, and transcutaneous electrical nerve stimulation on chronic tinnitus: A randomized controlled trial. Integr. Med. Res. 2020, 9, 100409. [Google Scholar] [CrossRef] [PubMed]
  31. Kim, B.H.; Kim, K.; Nam, H.J. A comparative study on the effects of systemic manual acupuncture, periauricular electroacupuncture, and digital electroacupuncture to treat tinnitus: A randomized, paralleled, open-labeled exploratory trial. BMC Complement. Altern. Med. 2017, 17, 1–7. [Google Scholar] [CrossRef]
  32. Yakunina, N.; Kim, S.S.; Nam, E.-C. BOLD fMRI effects of transcutaneous vagus nerve stimulation in patients with chronic tinnitus. PLoS ONE 2018, 13, e0207281. [Google Scholar] [CrossRef]
  33. Hyvärinen, P.; Yrttiaho, S.; Lehtimäki, J.; Ilmoniemi, R.; Mäkitie, A.; Ylikoski, J.; Mäkelä, J.P.; Aarnisalo, A.A. Transcutaneous Vagus Nerve Stimulation Modulates Tinnitus-Related Beta- and Gamma-Band Activity. Ear Hear. 2015, 36, e76–e85. [Google Scholar] [CrossRef]
  34. Ylikoski, J.; Markkanen, M.; Pirvola, U.; Lehtimäki, J.A.; Ylikoski, M.; Jing, Z.; Sinkkonen, S.T.; Mäkitie, A. Stress and Tinnitus; Transcutaneous Auricular Vagal Nerve Stimulation Attenuates Tinnitus-Triggered Stress Reaction. Front. Psychol. 2020, 11, 570196. [Google Scholar] [CrossRef] [PubMed]
  35. Peng, L.; Mu, K.; Liu, A.; Zhou, L.; Gao, Y.; Shenoy, I.T.; Mei, Z.; Chen, Q. Transauricular vagus nerve stimulation at auricular acupoints Kindey (CO10), Yidan (CO11), Liver (CO12) and Shenmen (TF4) can induce auditory and limbic cortices activation measured by fMRI. Hear. Res. 2018, 359, 1–12. [Google Scholar] [CrossRef]
  36. Peter, N.; Kleinjung, T. Neuromodulation for tinnitus treatment: An overview of invasive and non-invasive techniques. J. Zhejiang Univ. Sci. B 2018, 20, 116–130. [Google Scholar] [CrossRef] [PubMed]
  37. Collins, L.; Boddington, L.; Steffan, P.J.; McCormick, D. Vagus nerve stimulation induces widespread cortical and behavioral activation. Curr. Biol. 2021, 31, 2088–2098.e3. [Google Scholar] [CrossRef] [PubMed]
  38. Souza, R.R.; Robertson, N.M.; McIntyre, C.K.; Rennaker, R.L.; Hays, S.A.; Kilgard, M.P. Vagus nerve stimulation enhances fear extinction as an inverted-U function of stimulation intensity. Exp. Neurol. 2021, 341, 113718. [Google Scholar] [CrossRef]
  39. Sivaji, V.; Grasse, D.W.; Hays, S.A.; Bucksot, J.E.; Saini, R.; Kilgard, M.P.; Rennaker, R.L. ReStore: A wireless peripheral nerve stimulation system. J. Neurosci. Methods 2019, 320, 26–36. [Google Scholar] [CrossRef]
  40. Ganzer, P.D.; Darrow, M.J.; Meyers, E.C.; Solorzano, B.R.; Ruiz, A.D.; Robertson, N.M.; Adcock, K.S.; James, J.T.; Jeong, H.S.; Becker, A.M.; et al. Closed-loop neuromodulation restores network connectivity and motor control after spinal cord injury. Elife 2018, 7, e32058. [Google Scholar] [CrossRef]
  41. Kilgard, M.P.; Rennaker, R.L.; Alexander, J.; Dawson, J. Vagus nerve stimulation paired with tactile training improved sensory function in a chronic stroke patient. Neurorehabilitation 2018, 42, 159–165. [Google Scholar] [CrossRef]
  42. Tyler, R.; Cacace, A.; Stocking, C.; Tarver, B.; Engineer, N.; Martin, J.; Deshpande, A.; Stecker, N.; Pereira, M.; Kilgard, M.; et al. Vagus Nerve Stimulation Paired with Tones for the Treatment of Tinnitus: A Prospective Randomized Double-blind Controlled Pilot Study in Humans. Sci. Rep. 2017, 7, 11960. [Google Scholar] [CrossRef]
  43. Stander, J.; Grimmer, K.; Brink, Y. Time as a barrier to evidence uptake—A qualitative exploration of the concept of time for clinical practice guideline uptake by physiotherapists. J. Eval. Clin. Pract. 2020, 27, 280–290. [Google Scholar] [CrossRef]
  44. Lehane, E.; Agreli, H.; Connor, S.O.; Hegarty, J.; Warren, P.L.; Bennett, D.; Blake, C.; Burke, F.; Corrigan, M.; Drennan, J.; et al. Building capacity: Getting evidence-based practice into healthcare professional curricula. BMJ Evidence-Based Med. 2021, 26, 246. [Google Scholar] [CrossRef]
  45. Worum, H.; Lillekroken, D.; Roaldsen, K.S.; Ahlsen, B.; Bergland, A. Physiotherapists’ perceptions of challenges facing evidence-based practice and the importance of environmental empowerment in fall prevention in the municipality-a qualitative study. BMC Geriatr. 2020, 20, 432. [Google Scholar] [CrossRef]
  46. Huang, K.; Liang, S.; Chen, L.; Grellet, A. Acupuncture for tinnitus: A systematic review and meta-analysis of randomized controlled trials. Acupunct. Med. 2021, 39, 264–271. [Google Scholar] [CrossRef]
  47. Zeng, F.-G.; Richardson, M.; Tran, P.; Lin, H.; Djalilian, H. Tinnitus Treatment Using Noninvasive and Minimally Invasive Electric Stimulation: Experimental Design and Feasibility. Trends Hear. 2019, 23, 2331216518821449. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram.
Figure 1. PRISMA flow diagram.
Jcm 12 03673 g001
Figure 2. Rob-2 Tool Cochrane. D1. Randomization process; D2. deviations from the intended interventions; D3. missing outcome data; D4. measurement of the outcome; D5. selection of the reported results.
Figure 2. Rob-2 Tool Cochrane. D1. Randomization process; D2. deviations from the intended interventions; D3. missing outcome data; D4. measurement of the outcome; D5. selection of the reported results.
Jcm 12 03673 g002
Figure 3. (A) Comparison of the effects of unilateral auricular stimulation vs. Control for THI. (B) Comparison of the effects of unilateral auricular stimulation vs. Control for VAS Loudness. (C) Comparison of the effects of unilateral auricular stimulation vs. Control for VAS Uncomfortable [29,30,31].
Figure 3. (A) Comparison of the effects of unilateral auricular stimulation vs. Control for THI. (B) Comparison of the effects of unilateral auricular stimulation vs. Control for VAS Loudness. (C) Comparison of the effects of unilateral auricular stimulation vs. Control for VAS Uncomfortable [29,30,31].
Jcm 12 03673 g003
Table 1. Summary of all included studies.
Table 1. Summary of all included studies.
Study (Design)Number of Participants &
Male/Female
Age (Years)Duration TinnitusIntervention Duration
(Sessions/Weeks)
Area of Treatment
Comparison and Outcome MeasureBetween−Groups Differences (95% CI) [hg]
Kim et al., 2020 [30] (RCT)G1; at-VNS; 15.
G2; MA; 15
G3; 15
13/2
12/3
8/7
47.3 ± 9.5
48.6 ± 7.8
46.2 ± 11.0
98.1 ± 97.7 months
105.1 ± 142.0 months
107.8 ± 115.9 months
G1; 15; 30-Hz
G2; MA
G3; 30/90 Hz with a 3-second interval)
20 min for 10 sessions
Inner tragus, cymba conchae
THI, VAS loudness, and
VAS annoyance
PTA, and speech discrimintion test
G1 vs. G2 vs. G3
THI
Post−treatment (−0.16, 1.3) [0.57]
Short term (−0.25, 1.2) [0.48]
VAS Loudness
Post−treatment (−032, 1.13) [0.41]
Short term (−0.53, 0.9) [0.18]
VAS Uncomfortable
Post−treatment (−0.45, 0.59) [0.07]
Short term (−0.36, 0.68) [0.16]
Tutar et al., 2019 [29] (RCT)G1; TENS (unilateral tinnitus); 20
G2; TENS (bilateral tinnitus); 20
G3; Control; 20
NR
NR
NR
41.17 ± 10.7531 ± 49 months [range: 3–198 months]G1 and G2; 1–200 Hz, 10–30 mA, 1000 msec
10 sessions in 1 month for 30 min (maximum of 4 days between
each session)
Inner tragus, cymba conchae
THI
DASS
(pure tone audiometry, tympanometry, speech discrimination
score
G1 vs. G2 vs. G3
THI
Post−treatment (0.11, 1.4) [0.76]
Yakunina et al., 2018 [32] (CT)G1; at-VNS with Tinnitus subjects; 36
G2; at-VNS with Normal subjects.; 37
27/9
18/19
51.0 ± 11.9
30.9 ± 8.2
63.2 ± 59.5 (3–134) monthsG1 & G2; 500 μs, 25 Hz, 0.1 mA
Unique session of MRI scan, total of six 5 min fMRI runs
Inner tragus, cymba conchae
Hearing and tinnitus assessment
MRI
NR
Kim et al., 2017 [31] (RCT)G1; at-VNS (auricular region); 14.
G2; MA; 13
G3; at-VNS (distal region), 15
12/2
6/6
10/3
54.6 ± 14.3
49.3 ± 15.6
53.9 ± 13.2
9.55 ± 11.55 years
7.1 ± 7.85 years
5.7 ± 6.4 years
G1 and G3; 4/100 Hz interval 3 s
20 min for 8 sessions
2 sessions/week
Inner tragus, cymba conchae
THIS,
VAS loudness, and VAS uncomfortable, PTA, and speech discrimintion test
G1 vs. G2 vs. G3G1 vs. G2
Function (IKDC function subscale)
G1 vs. G2
G1 vs. G2
THI
Post−treatment (−0.66, 0.85) [0.1]
Short Term (−0.64, 0.87) [0.12]
VAS Loudness
Post−treatment (−0.81, 0.7) [−0.06]
Short term (−0.69, 0.82) [0.06]
VAS Uncomfortable
Post−treatment (−0.88, 0.63) [0.25]
Short term (−0.69, 0.82) [0.06]
Hamilton et al., 2016 [28] (CT)G1; Bi-modal stimulation Tinnitus compliant; NR
G2; Bi-modal stimulation Tinnitus non-compliant; NR
34 (63%) men
47.5 ± 11>2 years/<2 years 36 (78%)/10 (22%)G1; Auditory stimulus G2; Bi-modal stimulation. 20 Hz—20 kHz
For 14 weeks every visit per 2 weeks, 7 sessions, 47 min each session
Inner tragus, cymba conchae
THI, MML, TLM, and VAS loudness
G1 vs. G2
THI
Post−treatment (−0.35, 0.93) [0.29]
DASS—Depression Anxiety Stress scale; THI—Tinnitus Handicap Index; TENS—transcutaneous electrical nerve stimulation; at-NVS—auricular transcutaneous Nerve Vagus stimulation; ABVN—auricular branch of the vagus nerve; MA—manual acupuncture; EA—electrical acupuncture; PTA—pure tone average;. SD—speech discrimination; HRV—heart rate variability; PE—periauricular electroacupuncture group; DE—distal electroacupuncture group; MML—minimum masking level; TLM—Tinnitus loudness matching; MEG—magnetoencephalography; fMRI—functional Magnetic Resonance Image; CT—clinical trial; RCT—randomized controlled trial.
Table 2. Score of randomized clinical trials with PEDro scale.
Table 2. Score of randomized clinical trials with PEDro scale.
12345678910Total
Tinnitus
Kim et al., 2020 [30]YYYNNNNYYY6/10
Tutar et al., 2019 [29]YNYNNYYNYY6/10
Kim et al., 2017 [31]YYYNNNYYYY7/10
Yakunina et al., 2018 [32]NNNNNNNNYY2/10
Hamilton et al., 2016 [28]NNNNNNNNYY2/10
1: Random Allocation of Participants; 2: Concealed Allocation; 3: Similarity Between Groups at Baseline; 4: Participant Blinding; 5: Therapist Blinding; 6: Assessor Blinding; 7: Fewer than 15% Dropouts; 8: Intention- to-Treat Analysis; 9: Between-Group Statistical Comparisons; 10: Point Measures and Variability Data.
Table 3. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) Evidence Profile.
Table 3. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) Evidence Profile.
Number of StudiesRisk of BiasInconsistencyIndirectness of EvidenceImprecisionPublication BiasQuality of Evidencehg [95% CI]
Unilateral auricular stimulation vs. Control THI
Post-treatment group (n = 3)SeriousHighLow/acceptableNoVery seriousVery Low0.69 (0.06, 1.32)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.28 (−0.25, 0.81)
Unilateral auricular stimulation vs. Control Vas Loudness
Post-treatment group (n = 2)NoLowLow/acceptableNoVery seriousModerate−0.01 (−0.47, 0.45)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate−0.02 (−0.48, 0.44)
Unilateral auricular stimulation vs. Control Vas Uncomfortable
Post-treatment group (n = 2)NoLowLow/acceptableNoVery seriousModerate0.31 (−0.34, 0.96)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.31 (−0.16, 0.77)
Unilateral Stimulations vs. Bilateral THI
Post-treatment group (n = 4)SeriousHighLow/acceptableNoVery seriousVery Low0.21 (−0.13, 0.55)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.3 (−0.22, 0.83)
Unilateral Stimulations vs. Bilateral Vas Loudness
Post-treatment group (n = 2)NoLowLow/acceptableNoVery seriousModerate0.19 (−0.34, 0.71)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.13 (−0.39, 0.65)
Unilateral Stimulations vs. Bilateral Vas Uncomfortable
Post-treatment group (n = 2)NoLowLow/acceptableNoVery seriousModerate0.15 (−0.38, 0.67)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.16 (−0.36, 0.67)
Tens vs. Placebo THI
Post-treatment group (n = 3)SeriousModeratelow/acceptableNoVery seriousVery Low0.51 (0.1, 0.91)
Short-term subgroup (n = 2)NoLowlow/acceptableNoVery seriousModerate0.3 (−0.23, 0.83)
Tens vs. Placebo Vas Loudness
Post-treatment group (n = 2)NoLowLow/acceptableNoVery seriousModerate0.19 (−0.34, 0.71)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.13 (−0.39, 0.65)
Tens vs. Placebo Vas Uncomfortable
Post-treatment group (n = 2)NoLowLow/acceptableNoVery seriousModerate0.07 (−0.45, 0−59)
Short-term subgroup (n = 2)NoLowLow/acceptableNoVery seriousModerate0.16 (−0.36, 0.68)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fernández-Hernando, D.; Fernández-de-las-Peñas, C.; Machado-Martín, A.; Angulo-Díaz-Parreño, S.; García-Esteo, F.J.; Mesa-Jiménez, J.A. Effects of Non-Invasive Neuromodulation of the Vagus Nerve for Management of Tinnitus: A Systematic Review with Meta-Analysis. J. Clin. Med. 2023, 12, 3673. https://doi.org/10.3390/jcm12113673

AMA Style

Fernández-Hernando D, Fernández-de-las-Peñas C, Machado-Martín A, Angulo-Díaz-Parreño S, García-Esteo FJ, Mesa-Jiménez JA. Effects of Non-Invasive Neuromodulation of the Vagus Nerve for Management of Tinnitus: A Systematic Review with Meta-Analysis. Journal of Clinical Medicine. 2023; 12(11):3673. https://doi.org/10.3390/jcm12113673

Chicago/Turabian Style

Fernández-Hernando, David, Cesar Fernández-de-las-Peñas, Ana Machado-Martín, Santiago Angulo-Díaz-Parreño, Francisco J. García-Esteo, and Juan A. Mesa-Jiménez. 2023. "Effects of Non-Invasive Neuromodulation of the Vagus Nerve for Management of Tinnitus: A Systematic Review with Meta-Analysis" Journal of Clinical Medicine 12, no. 11: 3673. https://doi.org/10.3390/jcm12113673

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

Article Metrics

Back to TopTop