v
Search
Advanced Search

Publications > Journals > Journal of Translational Gastroenterology > Article Full Text

  • OPEN ACCESS

Vagus Nerve Stimulation and Sacral Nerve Stimulation for Inflammatory Bowel Disease: A Systematic Review

  • Victor Pikov* 
 Author information
Journal of Translational Gastroenterology   2023;1(2):94-100

doi: 10.14218/JTG.2023.00098

Abstract

Background and objectives

In this systematic review, we assessed the efficacy, potential mechanisms, and safety of two neuromodulation therapies in patients with inflammatory bowel disease (IBD), including Crohn’s disease and ulcerative colitis. The first therapy is vagus nerve stimulation (VNS) utilizing implantable or transcutaneous electrodes, and the second is sacral nerve stimulation (SNS) using implantable or percutaneous electrodes.

Methods

We conducted a systematic literature review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The PubMed database was comprehensively searched, and studies were rigorously assessed for inclusion and exclusion criteria.

Results

Our analysis encompassed five clinical studies, three on VNS and two on SNS. Most investigated studies demonstrated significant beneficial effects on IBD symptoms, including disease activity, severity of intestinal lesions, and intestinal pain. When evaluating the impact on key IBD pathophysiologies, both VNS and SNS exhibited trends toward reducing biomarkers of intestinal mucosal inflammation and mitigating sympathetic dominance. Importantly, none of the evaluated neuromodulation methods resulted in long-term adverse effects.

Conclusions

Cumulative evidence from the evaluated studies indicates that VNS and SNS therapies effectively alleviate IBD symptoms and may hold promise in addressing the underlying pathophysiologies of IBD, including intestinal mucosal inflammation and sympathetic dominance. Consequently, they represent valuable options for individualized IBD treatment.

Keywords

Vagus nerve stimulation, Sacral nerve stimulation, Neuromodulation, Inflammatory bowel disease, Crohn’s disease, Ulcerative colitis

Introduction

The prevalence of IBD is approximately 0.9% of the general population in the US,1 comprising 0.4% with ulcerative colitis (UC) and 0.5% with Crohn’s disease (CD).2 The direct cost of IBD care in the US amounts to $60 billion ($23K per patient),3 which is mainly aimed at treating IBD symptoms, such as diarrhea, blood in the stool, weight loss, fever, and abdominal pain. The etiology of IBD is multifactorial, involving genetic predisposition and immunologic disturbances.4 An exaggerated mucosal immune response to the patient’s native microbiota is pivotal in initiating and perpetuating intestinal inflammation.5 The anti-inflammatory medications include first-line small-molecule drugs like aminosalicylates (e.g., mesalamine), corticosteroids (e.g., budesonide, prednisone), and immunosuppressants (e.g., azathioprine These are followed by second-line anti-inflammatory biologics, such as anti-TNF-α and anti-α4β7 integrin antibodies.6 However, biological treatments are primarily symptomatic, do not prevent the recurrence of flares, and are associated with significant side effects. Furthermore, approximately 50% of UC7 and CD patients8 do not initially respond favorably to these treatments. Additionally, within the first year of drug use, 30–35% of initial responders experience a secondary loss of clinical response.9–11 For patients with CD and UC who are refractory to biologic medications, irreversible surgical procedures may be necessary to remove inflamed portions of the intestine.12 Removal of the colon (colectomy) is associated with an 81% risk of postoperative complications (e.g., depression, work productivity, diet restrictions, body image, sexual function)13 and a high cost of $140K.14 Low adherence to drug self-administration is an unsolved need in IBD patients.15 Given the shortcomings of existing therapies, there is a clear need for more effective, patient-adherent, and less expensive strategies for IBD treatment.

Recently, the VNS and SNS methods have been evaluated in the animal models of IBD. Both methods reduced the levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-18). They increased the levels of anti-inflammatory cytokines (IL-10, TGF-β) in the blood plasma, indicating healing of intestinal mucosal inflammation.16–24 The effectiveness of the VNS and SNS in IBD could be explained by normalizing the sympathetic and parasympathetic signaling, as the autonomic balance is shifted toward sympathetic dominance during the flares.25–29 In the rodent models of IBD, the VNS and SNS therapies effectively reduced sympathetic dominance in the autonomic balance.16–18,30 The SNS effects on the autonomic balance are likely mediated via two neural pathways: direct efferent sacral pathway to the colon and indirect spinal afferent-vagal efferent pathway to the colon.31 The VNS and SNS are typically applied using a minimally invasive procedure by implanting the VNS electrodes on the cervical vagus32,33 or SNS electrodes in the sacral foramen.34 Both VNS and SNS neuromodulation procedures have received FDA approval for various indications, demonstrating a well-established safety profile during long-term implantation and use.35–38 In addition to the implantable VNS and SNS methods, two non-implantable approaches have also shown a safe track record during intermittent daily use: transcutaneous auricular vagus nerve stimulation39 and percutaneous sacral nerve stimulation based on electroacupuncture.40

In summary, despite the availability of various symptomatic treatments for IBD, there remains a gap in therapies that specifically target the underlying pathophysiologies of the disease. Two neuromodulation therapies, VNS and SNS, have been investigated as potential treatments for IBD. These therapies encompass both implantable and transcutaneous VNS, as well as implantable and percutaneous SNS. In this systematic review, we have assessed the effectiveness of these neuromodulation approaches in addressing IBD symptoms and key pathophysiologies, including intestinal mucosal inflammation and sympathetic dominance.

Materials and methods

The systematic review was performed per the 2020 version of the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) statement.41

Data sources and searches

On December 25, 2023, the PubMed database was searched for scientific papers. The search keywords are provided in Table 1.

Table 1

Search keywords for PubMed for two evaluated therapies

TherapySearch keywords for PubMed
VNS(“vagus nerve stimulation” [Title/Abstract] OR “vagal nerve stimulation” [Title/Abstract] OR (auricular[Title/Abstract] AND “nerve stimulation” [Title/Abstract])) AND (“Inflammatory Bowel Disease*” [Title/Abstract] OR “Crohn*” [Title/Abstract] OR colitis[Title/Abstract])
SNS(“sacral nerve stimulation” [Title/Abstract] OR “sacral neuromodulation” [Title/Abstract]) AND (“Inflammatory Bowel Disease*” [Title/Abstract] OR “Crohn*” [Title/Abstract] OR colitis[Title/Abstract])

Data collection and evaluation

All identified records were imported into the EPPI Reviewer Software for review. Abstracts were initially screened, followed by full text reviewed per the inclusion and exclusion criteria.

Study selection

Both sham-controlled RCTs and open-label single-arm non-RCTs were considered for inclusion in our review. Due to the invasive nature of implantable VNS and SNS therapies, the inclusion of a sham group was deemed unethical. The criteria for inclusion were as follows: (1) papers published in English, (2) papers with full-text availability, and (3) presentation of statistical results. Animal studies, non-English papers, reviews, abstracts, and case studies were excluded from consideration.

Results

Search outcomes

For the VNS and SNS searches, we initially identified 69 and 27 records, respectively, from the PubMed database, which were then imported into the EPPI Reviewer software.42 Upon screening, all records were successfully retrieved and assessed for eligibility. Among the 89 records excluded, 64 were related to VNS, and 25 were related to SNS. The exclusions comprised 51 reviews (39 VNS and 12 SNS), 27 animal studies (19 VNS and 8 SNS), 7 records without an assessment of IBD symptoms (2 VNS and 5 SNS), 1 case study (VNS), 1 abstract (VNS), 1 study protocol (VNS), and 1 record unrelated to VNS therapy.

Subsequently, we included 7 records in the analysis: 5 for VNS and 2 for SNS. A visual representation of the record selection process is depicted in Figure 1 using the PRISMA flow diagram. The results of the VNS and SNS studies are summarized in Tables 2 and 3,43–49 respectively. These tables present the IBD symptom outcomes as the mean ± standard deviation of the difference between post-therapy data and pre-therapy data.

PRISMA flow diagram of record selection.
Fig. 1  PRISMA flow diagram of record selection.

IBD, inflammatory bowel disease; PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses; SNS, sacral nerve stimulation; VNS, vagus nerve stimulation.

Table 2

Summary of three clinical studies evaluating the effects of VNS therapy on CD and UC

NCT number, durationN per armDaily therapy doseCDAI, pCDAI, pUCDAICDEISGI-related VASCRP (mg/l)FC (µg/g)LF/HFRef.
NCT01569503, 6 months5 CD2.2 h (24 h * 9%)Δ −166 ± 58, p < 0.01Δ −6.8 ± 1.5, p < 0.001Δ −2.1 ± 1.4, p < 0.05Δ −3.6 ± 5.7, p > 0.05Δ −1,070 ± 1,301, p > 0.05Δ −2.4 ± 2.3, p > 0.0543,44
NCT01569503, 12 months7 CD2.2 h (24 h 9%)Δ −156 ± 62, p < 0.001Δ −4.2 ± 4.1, p < 0.05Δ −1.9 ± 1.9, p < 0.05Δ −7.9 ± 9.3, p > 0.05Δ −1,168 ± 912, p > 0.05Δ −1.7 ± 2, p > 0.0545
NCT02311660, 4 months12 CD20 min (5 min * 4x)Δ −115 ± 24, p < 0.001NDNDΔ −0.9 ± 0.9, p > 0.05Δ −3,209 ± 937, p < 0.01ND46
NCT03863704, 3.5 months10 CD10 min (5 min * 2x)Δ −15 ± 17, p < 0.05NDNDNDΔ −357 ± 800, p > 0.05ND47
NCT03863704, 3.5 months12 UC10 min (5 min * 2x)Δ −8 ± 15, p > 0.05NDNDNDΔ −833 ± 250, p < 0.05ND47
Table 3

Summary of two clinical studies evaluating the effects of SNS and sham therapies

NCT number, durationN per armDaily therapy doseUCDAIMayo scoreTNF-α (pg/ml)CRP (mg/l)FC (µg/g)LF/HFRef.
NCT02748590, 4 months8 UC + SNS24 hΔ −1.5 ± 2.8, p > 0.05NDNDNDΔ −234 ± NDND48
N/A, 2 weeks15 UC + SNS6 min (60 min *10%)NDΔ −2.1 ± 1.8, p < 0.01Δ −9.5 ± 8.9, p < 0.001Δ −6.6 ± 9.9, p < 0.05NDΔ −0.68 ± 1.78, p > 0.0549
N/A, 2 weeks11 UC + sham6 min (60 min *10%)NDΔ −0.6 ± 1.3, p > 0.05Δ −5.4 ± 14.1, p > 0.05Δ −1.0 ± 6.7, p > 0.05NDΔ 0.96 ± 1.13, p < 0.0549

VNS studies

We identified three VNS studies, including two open-label single-arm iVNS studies (with VNS electrodes implanted on the cervical vagus) and one RCT taVNS study (with transcutaneous auricular VNS electrodes).

In two iVNS studies, NCT01569503 and NCT02311660,43–46 bipolar electrical stimulation was delivered via the helical cuff electrode (Model 302 or 304, Cyberonics) implanted on the left cervical vagus nerve and tunneled to an IPG (Model 102 or 103, Cyberonics), which was placed in the subcutaneous pocket on the left chest wall. The first iVNS study (NCT01569503) was performed in Grenoble, France, where 7 adult patients with moderate CD (220 ≤ CDAI ≤ 450) diagnosed for at least 3 months before enrollment and naive of biologic treatment were subjected to iVNS with the following parameters: frequency of 10 Hz, current amplitude of 0.5–1.25 mA, pulse width of 500 µs, duty cycle of 9% (0.5 min every 5.5 min), delivered continuously for 12 months.43–45 The second iVNS study (NCT02311660) was performed at four European locations (Zagreb, Croatia; Milano, Italy; Amsterdam, Netherlands; and Stockholm, Sweden), where adult patients with moderate CD (220 ≤ CDAI ≤ 450, SES-CD ≥ 2 in at least one segment, and FC ≥ 200 ug/g) diagnosed for at least 4 months before enrollment and refractory or intolerant to at least one biologic treatment (infliximab, adalimumab, or vedolizumab) were subjected to iVNS with the following parameters: frequency of 10 Hz, current amplitude of 0.25–2.0 mA, pulse width of 250 µs, duty cycle of 100%, delivered for 5 minutes four times per day for 4 months.46

In the taVNS study NCT03863704,47 active bipolar electrical stimulation was delivered via the hand-held skin probe with two electrodes (Blue Moon Health) placed on the cymba concha area inside the left ear, while sham electrical stimulation was delivered in the middle of the left calf, with the cross-over design, where patients served as their controls. The study was performed in New York, USA, where pediatric and young adult patients (10–21 years) with mild and moderate CD and UC (FC ≥ 200 ug/g) diagnosed for at least 3 months prior to enrollment and irrespective of biologic treatment (only those on infliximab were excluded) were subjected to taVNS with the following parameters: frequency of 20 Hz, current amplitude just below the pain threshold, pulse width of 300 µs, duty cycle of 100%, delivered for 5 minutes two times per day for 3.5 months.47

The results of three VNS studies are summarized in Table 2.

The clinical studies examined the long-term effects of iVNS and taVNS on IBD symptoms, with a follow-up period of up to 12 months for iVNS and 3.5 months for taVNS.

In all iVNS and taVNS studies, a significant reduction in IBD disease activity was observed in CD patients. This reduction was assessed using the CDAI and GI-related VAS in adults and the pCDAI in adolescents. However, this reduction was not observed in adolescent patients with UC who were treated with taVNS, as assessed by the pUCDAI.

One of the iVNS studies (NCT01569503) also demonstrated a significant long-term improvement, both at 6 and 12 months, in the severity of intestinal lesions, assessed endoscopically as the CDEIS, as well as in intestinal pain, assessed using the GI-related VAS.

Two iVNS studies (NCT01569503, NCT02311660) additionally assessed blood biomarkers, including CRP, and fecal biomarkers, including FC, for intestinal mucosal inflammation. In both iVNS studies, CRP was not significantly reduced in CD patients. However, FC was significantly reduced in only one of the two studies (NCT02311660).

Regarding the taVNS study, FC was assessed, but the results were inconclusive. FC was significantly reduced in UC patients but not in CD patients.

One of two iVNS studies (NCT01569503) evaluated a possible mechanism of action for the VNS therapy, the recovery of the autonomic balance (assessed as the LF/HF ratio of the power spectrum of the HRV derived from the electrocardiogram) and, while there was a trend toward a decreased sympathetic dominance at both 6 and 12 months, it was not statistically significant.

The iVNS therapy is associated with the risk of surgical and post-surgical complications. Among 16 implanted subjects in the NCT02311660 study, one experienced transient postoperative skin infection requiring device explantation. No complications were reported in the NCT01569503 study. In both iVNS studies, the VNS-related adverse effects included only discomfort due to voice hoarseness, a typical iVNS side effect. In the taVNS study (NCT03863704), one subject developed a transient skin redness and a minor break in the skin because of excessive pressure applied to the ear with the taVNS probe during the first week of stimulation, which was resolved by further educating the subject on the taVNS technique.

SNS studies

We identified two studies related to SNS: one open-label single-arm study involving implantable SNS electrodes (iSNS) and one RCT focusing on percutaneous SNS electrodes (pSNS).

In the iSNS study, registered as NCT02748590,48 bipolar electrical stimulation was administered through a 4-electrode SNS lead (Model 3889, Medtronic) implanted within the S3 foramen. The lead was then tunneled to connect with an IPG, specifically the InterStim II (Model 3058, Medtronic), placed in a subcutaneous pocket on the left chest wall. This study was conducted in Nantes, France. It involved eight adult patients diagnosed with moderate ulcerative UC who exhibited a UCDAI ranging from 6 to 9 and an endoscopic UCDAI score of at least 2. These patients had been diagnosed with UC for at least 2 years before enrollment and were resistant to immunosuppressive or biologic anti-TNF treatments. During the iSNS intervention, the following stimulation parameters were employed: a frequency of 14 Hz, a current amplitude of 1.1 V, a pulse width of 210 µs, a duty cycle of 100%, and continuous stimulation for a duration of 4 months.48

In the pSNS study,49 bipolar electrical stimulation was delivered via four stainless steel acupuncture needles (diameter 0.45 mm, length 100–125 mm) inserted bilaterally inside the S3 and S4 foramens and attached to an external stimulator (Transcutaneous Electrical Applicator, Model SNM-FDC01, MedKinetic Medical Device Co. Ltd, Ningbo, China), while the sham electrical stimulation was delivered using the same needles placed 20 mm downward and 8–10 cm lateral from these sacral foramina. The study was performed in Nanjing, China, where 26 adult patients with mild and moderate UC (3 ≤ Mayo score ≤ 10) diagnosed for at least 3 months prior to enrollment were subjected to pSNS with the following parameters: frequency of 5 Hz, current amplitude of 2–10 mA, pulse width of 500 µs, duty cycle of 10% (10 sec every 100 sec), delivered for 1 hour per day for 2 weeks.49

The results of two SNS studies are summarized in Table 3.

The clinical studies evaluated the medium-term effect of iSNS and pSNS on IBD symptoms with a follow-up period of 4 months for iSNS and 2 weeks for pSNS. Only the UC patients were evaluated in both studies: in the iSNS study, the UC disease activity assessed as the UCDAI was insignificantly reduced, while in the pSNS study, it was assessed as the Mayo score and was significantly reduced in the SNS arm but not in the sham arm. The pSNS study also assessed the blood biomarkers of intestinal mucosal inflammation (TNF-α and CRP), with both biomarkers significantly reduced in the SNS arm but not in the sham arm. In contrast, the iSNS study assessed the fecal biomarker FC, and while the FC level was reduced post-SNS, no statistical significance calculation was provided for that effect.

The pSNS study also evaluated a possible mechanism of action for the SNS therapy, the recovery of the autonomic balance (assessed as the LF/HF ratio). While there was a statistically insignificant trend toward a decreased sympathetic dominance in the SNS arm, the sympathetic dominance significantly worsened in the sham arm.

The iSNS therapy is associated with the risk of surgical and post-surgical complications. Among 8 implanted subjects in the NCT02748590 study, one lead disconnection occurred during the test phase, while the implanted lead was percutaneously connected to the external stimulator. There were no SNS-related adverse effects in both the iSNS and pSNS studies.

Discussion

This systematic review assessed the efficacy, mechanisms, and safety of two parasympathetic neuromodulation methods (VNS and SNS) in patients with IBD. Notably, all VNS treatment arms involving patients with CD and the pSNS treatment arm involving patients with UC demonstrated significant and beneficial neuromodulatory effects on IBD symptoms. These effects encompassed improvements in disease activity, the severity of intestinal lesions, and alleviation of intestinal pain.

However, a contrast was observed in the case of iSNS, involving eight UC patients, and pSNS in eleven CD patients. These treatments showed insignificant trends in improving IBD symptoms, which could be attributed to the relatively small number of patients and/or the rather short follow-up duration, with the pSNS study spanning only 2 weeks.

Furthermore, evaluating blood and fecal biomarkers related to intestinal mucosal inflammation in the VNS and SNS studies yielded inconclusive results. In the VNS studies, these biomarker changes did not demonstrate clear trends, while in the pSNS study, blood biomarkers such as TNF-α and CRP significantly reduced in the pSNS treatment arm, contrasting with a lack of effect observed in the sham pSNS arm.

The potential mechanism of action involving the recovery of autonomic balance was explored in one iVNS study (NCT01569503) and the pSNS study. These investigations showed a statistically insignificant trend toward a reduction in sympathetic dominance in both studies.

Neither of the evaluated neuromodulation methods was associated with long-term adverse effects. Both iVNS and iSNS can be considered safe for long-term use, while taVNS and pSNS provide even safer options for short-term treatment. The selection of an implanted vs. transcutaneous vs. percutaneous option would, therefore, depend on the patient’s risk acceptance and desired convenience, as transcutaneous vs. percutaneous options require daily placement of the stimulation electrodes.

Comparing and contrasting these neuromodulation methods for IBD treatment, several advantages and disadvantages are associated with each method, outlined in Table 4.

Table 4

Summary of advantages and disadvantages of VNS and SNS for IBD treatment

Neuromodulation methodAdvantagesDisadvantages
iVNS and iSNSConfirmed efficacy for treating CD (iVNS); “Implant it and forget it” approach of achieving high patient adherence for timely therapy useHigh cost and extensive clinical resources involved in the implantation procedure; Risks associated with the surgery and long-term implantation; Not FDA approved for treating IBD
taVNS and pSNSConfirmed efficacy for treating CD (taVNS) and UC (pSNS); Non-invasive; Inexpensive; Can be used at homeRequires the patient to learn the location of cymba concha area inside the ear (taVNS); Requires visits to an acupuncturist (pSNS); Not FDA approved for treating IBD

Additional clinical studies must be conducted to further evaluate the efficacy of the VNS and SNS methods. Based on the evaluated studies, both methods improve IBD symptoms and intestinal mucosal inflammation biomarkers. In addition, there is a trend toward decreased sympathetic dominance in both methods. In contrast, existing medications aim only for symptom alleviation without beneficial effects on sympathetic dominance.

The availability of VNS and SNS therapies provides clinicians with valuable options for optimizing the treatment of IBD patients. For example, the taVNS and/or pSNS may be prescribed as adjunct therapies to the anti-inflammatory medications, as their combined use may lead to longer-lasting and more sustainable results with fewer side effects. As the IBD severity worsens over time, the use of reversible and minimally-invasive iVNS and/or iSNS can also replace or delay the need for irreversible surgical procedures to resect an inflamed portion of the intestine, such as colectomy.

The study limitation is excluding studies published in other languages, while all relevant studies published in English have been included.

Conclusions

While the current clinical evidence does not yet firmly establish the role of the examined VNS and SNS therapies in directly targeting the IBD pathophysiologies, both therapies effectively alleviate IBD symptoms and impact biomarkers associated with intestinal mucosal inflammation. Consequently, they represent valuable options for personalized IBD treatment.

Given the relatively small sample sizes of the studies we have reviewed, larger-scale investigations in the future will be essential to arrive at definitive conclusions regarding the efficacy and mechanisms of action of VNS and SNS therapies in the context of IBD.

Abbreviations

CD: 

Crohn’s disease

CDAI: 

Crohn’s Disease activity index

pCDAI: 

pediatric CDAI

CDEIS: 

Crohn’s Disease endoscopic index of severity

CRP: 

C-reactive protein

FC: 

fecal calprotectin

FDA: 

Food and Drug Administration

GI: 

gastrointestinal

HRV: 

heart rate variability

IBD: 

inflammatory bowel disease

IPG: 

implantable pulse generator

iSNS: 

implantable SNS

iVNS: 

implantable VNS

LF/HF: 

low-frequency to high-frequency ratio

pSNS: 

percutaneous SNS using electroacupuncture needles

RCT: 

randomized controlled trial

SES-CD: 

simple endoscopic score for Crohn’s Disease

SNS: 

sacral nerve stimulation

taVNS: 

transcutaneous auricular VNS

TNF-α: 

Tumor Necrosis Factor-α

UC: 

ulcerative colitis

UCDAI: 

ulcerative colitis disease activity index

pUCDAI: 

pediatric UCDAI

VAS: 

Visual Analogue Scale

VNS: 

vagus nerve stimulation

Declarations

Acknowledgement

None.

Funding

Victor Pikov’s effort in preparing the manuscript was partially supported by a grant from the National Institutes of Health (U41NS129514).

Conflict of interest

Victor Pikov is the Founder and CEO of Medipace Inc.

References

  1. Dahlhamer JM, Zammitti EP, Ward BW, Wheaton AG, Croft JB. Prevalence of Inflammatory Bowel Disease Among Adults Aged ≥18 Years - United States, 2015. MMWR Morb Mortal Wkly Rep 2016;65(42):1166-1169 View Article PubMed/NCBI
  2. Sheriff MZ, Mansoor E, Luther J, Ananthakrishnan AN, Abou Saleh M, Ho E, et al. Opportunistic Infections Are More Prevalent in Crohn’s Disease and Ulcerative Colitis: A Large Population-Based Study. Inflamm Bowel Dis 2020;26(2):291-300 View Article PubMed/NCBI
  3. Park KT, Ehrlich OG, Allen JI, Meadows P, Szigethy EM, Henrichsen K, et al. The Cost of Inflammatory Bowel Disease: An Initiative From the Crohn’s & Colitis Foundation. Inflamm Bowel Dis 2020;26(1):1-10 View Article PubMed/NCBI
  4. Lovasz BD, Golovics PA, Vegh Z, Lakatos PL. New trends in inflammatory bowel disease epidemiology and disease course in Eastern Europe. Dig Liver Dis 2013;45(4):269-276 View Article PubMed/NCBI
  5. Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology 2011;140(6):1729-1737 View Article PubMed/NCBI
  6. Takatsu N, Hisabe T, Higashi D, Ueki T, Matsui T. Vedolizumab in the Treatment of Ulcerative Colitis: An Evidence-Based Review of Safety, Efficacy, and Place of Therapy. Core Evid 2020;15:7-20 View Article PubMed/NCBI
  7. Nguyen NH, Singh S, Sandborn WJ. Positioning Therapies in the Management of Crohn’s Disease. Clin Gastroenterol Hepatol 2020;18(6):1268-1279 View Article PubMed/NCBI
  8. Welty M, Mesana L, Padhiar A, Naessens D, Diels J, van Sanden S, et al. Efficacy of ustekinumab vs. advanced therapies for the treatment of moderately to severely active ulcerative colitis: a systematic review and network meta-analysis. Curr Med Res Opin 2020;36(4):595-606 View Article PubMed/NCBI
  9. Nishida Y, Hosomi S, Yamagami H, Yukawa T, Otani K, Nagami Y, et al. Neutrophil-to-Lymphocyte Ratio for Predicting Loss of Response to Infliximab in Ulcerative Colitis. PLoS One 2017;12(1):e0169845 View Article PubMed/NCBI
  10. Sands BE, Peyrin-Biroulet L, Loftus EV, Danese S, Colombel JF, Törüner M, et al. Vedolizumab versus Adalimumab for Moderate-to-Severe Ulcerative Colitis. N Engl J Med 2019;381(13):1215-1226 View Article PubMed/NCBI
  11. Qiu Y, Chen BL, Mao R, Zhang SH, He Y, Zeng ZR, et al. Systematic review with meta-analysis: loss of response and requirement of anti-TNFα dose intensification in Crohn’s disease. J Gastroenterol 2017;52(5):535-554 View Article PubMed/NCBI
  12. Lightner AL, Holubar SD. Yamada’s Textbook of Gastroenterology. ; 2022, 1324-1353 View Article
  13. Brown C, Gibson PR, Hart A, Kaplan GG, Kachroo S, Ding Q, et al. Long-term outcomes of colectomy surgery among patients with ulcerative colitis. Springerplus 2015;4:573 View Article PubMed/NCBI
  14. Ghoz H, Kesler A, Hoogenboom SA, Gavi F, Brahmbhatt B, Cangemi J, et al. Decreasing Colectomy Rates in Ulcerative Colitis in the Past Decade: Improved Disease Control?. J Gastrointest Surg 2020;24(2):270-277 View Article PubMed/NCBI
  15. Singer J, Rai V, Sossenheimer P, Karpin J, Rodriguez T, Yi Y, et al. P091 Factors Predicting Compliance With Passive and Active Monitoring in Inflammatory Bowel Disease Patients. Am. J. Gastroenterol 2019;114:S24 View Article
  16. Tu L, Gharibani P, Yin J, Chen JDZ. Sacral nerve stimulation ameliorates colonic barrier functions in a rodent model of colitis. Neurogastroenterol Motil 2020;32(10):e13916 View Article PubMed/NCBI
  17. Pasricha TS, Zhang H, Zhang N, Chen JDZ. Sacral nerve stimulation prompts vagally-mediated amelioration of rodent colitis. Physiol Rep 2020;8(1):e14294 View Article PubMed/NCBI
  18. Jin H, Guo J, Liu J, Lyu B, Foreman RD, Yin J, et al. Anti-inflammatory effects and mechanisms of vagal nerve stimulation combined with electroacupuncture in a rodent model of TNBS-induced colitis. Am J Physiol Gastrointest Liver Physiol 2017;313(3):G192-G202 View Article PubMed/NCBI
  19. Zhang N, Zhang H, Jiang L, Zhang S, Yin J, Schramm L, et al. A novel method of sacral nerve stimulation for colonic inflammation. Neurogastroenterol Motil 2020;32(6):e13825 View Article PubMed/NCBI
  20. Guo J, Jin H, Shi Z, Yin J, Pasricha T, Chen JDZ. Sacral nerve stimulation improves colonic inflammation mediated by autonomic-inflammatory cytokine mechanism in rats. Neurogastroenterol Motil 2019;31(10):e13676 View Article PubMed/NCBI
  21. Cheng J, Shen H, Chowdhury R, Abdi T, Selaru F, Chen JDZ. Potential of Electrical Neuromodulation for Inflammatory Bowel Disease. Inflamm Bowel Dis 2020;26(8):1119-1130 View Article PubMed/NCBI
  22. Brégeon J, Coron E, Da Silva AC, Jaulin J, Aubert P, Chevalier J, et al. Sacral nerve stimulation enhances early intestinal mucosal repair following mucosal injury in a pig model. J Physiol 2016;594(15):4309-4323 View Article PubMed/NCBI
  23. Meregnani J, Clarençon D, Vivier M, Peinnequin A, Mouret C, Sinniger V, et al. Anti-inflammatory effect of vagus nerve stimulation in a rat model of inflammatory bowel disease. Auton Neurosci 2011;160(1-2):82-89 View Article PubMed/NCBI
  24. Meroni E, Stakenborg N, Gomez-Pinilla PJ, De Hertogh G, Goverse G, Matteoli G, et al. Functional characterization of oxazolone-induced colitis and survival improvement by vagus nerve stimulation. PLoS One 2018;13(5):e0197487 View Article PubMed/NCBI
  25. Kim KN, Yao Y, Ju SY. Heart rate variability and inflammatory bowel disease in humans: A systematic review and meta-analysis. Medicine (Baltimore) 2020;99(48):e23430 View Article PubMed/NCBI
  26. Hirten RP, Danieletto M, Scheel R, Shervey M, Ji J, Hu L, et al. Longitudinal Autonomic Nervous System Measures Correlate With Stress and Ulcerative Colitis Disease Activity and Predict Flare. Inflamm Bowel Dis 2021;27(10):1576-1584 View Article PubMed/NCBI
  27. Ganguli SC, Kamath MV, Redmond K, Chen Y, Irvine EJ, Collins SM, et al. A comparison of autonomic function in patients with inflammatory bowel disease and in healthy controls. Neurogastroenterol Motil 2007;19(12):961-967 View Article PubMed/NCBI
  28. Gunterberg V, Simrén M, Öhman L, Friberg P, Jones MP, Van Oudenhove L, et al. Autonomic nervous system function predicts the inflammatory response over three years in newly diagnosed ulcerative colitis patients. Neurogastroenterol Motil 2016;28(11):1655-1662 View Article PubMed/NCBI
  29. Maunder RG, Greenberg GR, Nolan RP, Lancee WJ, Steinhart AH, Hunter JJ. Autonomic response to standardized stress predicts subsequent disease activity in ulcerative colitis. Eur J Gastroenterol Hepatol 2006;18(4):413-420 View Article PubMed/NCBI
  30. Sun P, Zhou K, Wang S, Li P, Chen S, Lin G, et al. Involvement of MAPK/NF-κB signaling in the activation of the cholinergic anti-inflammatory pathway in experimental colitis by chronic vagus nerve stimulation. PLoS One 2013;8(8):e69424 View Article PubMed/NCBI
  31. Tu L, Gharibani P, Zhang N, Yin J, Chen JD. Anti-inflammatory effects of sacral nerve stimulation: a novel spinal afferent and vagal efferent pathway. Am J Physiol Gastrointest Liver Physiol 2020;318(4):G624-G634 View Article PubMed/NCBI
  32. Chiba R, Enatsu R, Ochi S, Yamada S, Sasagawa A, Suzuki H, et al. Intraoperative Monitoring for Vagus Nerve Stimulation. World Neurosurg 2019;131:191-193 View Article PubMed/NCBI
  33. Kucia K, Merk W, Zapalowicz K, Medrala T. Vagus Nerve Stimulation For Treatment Resistant Depression: Case Series Of Six Patients - Retrospective Efficacy And Safety Observation After One Year Follow Up. Neuropsychiatr Dis Treat 2019;15:3247-3254 View Article PubMed/NCBI
  34. Banakhar M, Hassouna M. Patients with Sacral Neuromodulation: What Are the Factors Affecting Their Therapy Satisfaction?. Urol Int 2019;103(4):450-453 View Article PubMed/NCBI
  35. Siegel S, Noblett K, Mangel J, Griebling TL, Sutherland SE, Bird ET, et al. Three-year Follow-up Results of a Prospective, Multicenter Study in Overactive Bladder Subjects Treated With Sacral Neuromodulation. Urology 2016;94:57-63 View Article PubMed/NCBI
  36. Bielefeldt K. Adverse events of sacral neuromodulation for fecal incontinence reported to the federal drug administration. World J Gastrointest Pharmacol Ther 2016;7(2):294-305 View Article PubMed/NCBI
  37. Lim YG, Ker JRX, Tan YL, Chan DWS, Low DCY, Ng WH, et al. Adverse Events and Complications Associated With Vagal Nerve Stimulation: An Analysis of the Manufacturer And User Facility Device Experience Database. Neuromodulation 2023 View Article PubMed/NCBI
  38. Toffa DH, Touma L, El Meskine T, Bouthillier A, Nguyen DK. Learnings from 30 years of reported efficacy and safety of vagus nerve stimulation (VNS) for epilepsy treatment: A critical review. Seizure 2020;83:104-123 View Article PubMed/NCBI
  39. Kim AY, Marduy A, de Melo PS, Gianlorenco AC, Kim CK, Choi H, et al. Safety of transcutaneous auricular vagus nerve stimulation (taVNS): a systematic review and meta-analysis. Sci Rep 2022;12(1):22055 View Article PubMed/NCBI
  40. Chen S, Wang S, Xuan L, Lu H, Hu Z, Zhang C, et al. Comparison of efficacy and safety between electroacupuncture at ‘four sacral points’ and conventional electroacupuncture for the treatment of urinary incontinence after stroke: study protocol for a randomised controlled trial. BMJ Open 2018;8(11):e021783 View Article PubMed/NCBI
  41. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Int J Surg 2021;88:105906 View Article PubMed/NCBI
  42. Thomas J, Graziosi S, Brunton J, Ghouze Z, O’Driscoll P, Bond M. EPPI-Centre Software. London: UCL Social Research Institute; 2020
  43. Bonaz B, Sinniger V, Hoffmann D, Clarençon D, Mathieu N, Dantzer C, et al. Chronic vagus nerve stimulation in Crohn’s disease: a 6-month follow-up pilot study. Neurogastroenterol Motil 2016;28(6):948-953 View Article PubMed/NCBI
  44. Bonaz B, Sinniger V, Pellissier S. Vagus nerve stimulation: a new promising therapeutic tool in inflammatory bowel disease. J Intern Med 2017;282(1):46-63 View Article PubMed/NCBI
  45. Sinniger V, Pellissier S, Fauvelle F, Trocmé C, Hoffmann D, Vercueil L, et al. A 12-month pilot study outcomes of vagus nerve stimulation in Crohn’s disease. Neurogastroenterol Motil 2020;32(10):e13911 View Article PubMed/NCBI
  46. D’Haens G, Eberhardson M, Cabrijan Z, Danese S, van den Berg R, Löwenberg M, et al. Neuroimmune modulation through vagus nerve stimulation reduces inflammatory activity in Crohn’s disease patients: a prospective open label study. J Crohns Colitis 2023 View Article PubMed/NCBI
  47. Sahn B, Pascuma K, Kohn N, Tracey KJ, Markowitz JF. Transcutaneous auricular vagus nerve stimulation attenuates inflammatory bowel disease in children: a proof-of-concept clinical trial. Bioelectron Med 2023;9(1):23 View Article PubMed/NCBI
  48. Drissi F, Bourreille A, Neunlist M, Meurette G. Sacral neuromodulation for refractory ulcerative colitis: safety and efficacy in a prospective observational series of eight patients. Tech Coloproctol 2023;27(6):501-505 View Article PubMed/NCBI
  49. Chen Z, Li J, Ma Q, Pikov V, Li M, Wang L, et al. Anti-Inflammatory Effects of Two-Week Sacral Nerve Stimulation Therapy in Patients With Ulcerative Colitis. Neuromodulation 2023 View Article PubMed/NCBI
  • Journal of Translational Gastroenterology
  • eISSN 2994-8754
Back to Top

Vagus Nerve Stimulation and Sacral Nerve Stimulation for Inflammatory Bowel Disease: A Systematic Review

Victor Pikov
  • Reset Zoom
  • Download TIFF