Cervical transcutaneous vagal neuromodulation in chronic pancreatitis patients with chronic pain: A randomised sham controlled clinical trial

Janusiya Anajan Muthulingam, Søren Schou Olesen, Tine Maria Hansen, Christina Brock, Asbjørn Mohr Drewes, Jens Brøndum Frøkjær, Janusiya Anajan Muthulingam, Søren Schou Olesen, Tine Maria Hansen, Christina Brock, Asbjørn Mohr Drewes, Jens Brøndum Frøkjær

Abstract

Background & aims: Chronic abdominal pain is the primary symptom of chronic pancreatitis, but unfortunately it is difficult to treat. Vagal nerve stimulation studies have provided evidence of anti-nociceptive effect in several chronic pain conditions. We investigated the pain-relieving effects of transcutaneous vagal nerve stimulation in comparison to sham treatment in chronic pancreatitis patients.

Methods: We conducted a randomised double-blinded, sham-controlled, crossover trial in patients with chronic pancreatitis. Patients were randomly assigned to receive a two-week period of cervical transcutaneous vagal nerve stimulation using the gammaCore device followed by a two-week sham stimulation, or vice versa. We measured clinical and experimental endpoints before and after each treatment. The primary clinical endpoint was pain relief, documented in a pain diary using a visual analogue scale. Secondary clinical endpoints included Patients' Global Impression of Change score, quality of life and Brief Pain Inventory questionnaire. Secondary experimental endpoints included cardiac vagal tone and heart rate.

Results: No differences in pain scores were seen in response to two weeks transcutaneous vagal nerve stimulation as compared to sham treatment (difference in average pain score (visual analogue scale): 0.17, 95%CI (-0.86;1.20), P = 0.7). Similarly, no differences were seen for secondary clinical endpoints, except from an increase in the appetite loss score (13.9, 95%CI (0.5:27.3), P = 0.04). However, improvements in maximum pain scores were seen for transcutaneous vagal nerve stimulation and sham treatments as compared to their respective baselines: vagal nerve stimulation (-1.3±1.7, 95%CI (-2.21:-0.42), P = 0.007), sham (-1.3±1.9, 95%CI (-2.28:-0.25), P = 0.018). Finally, heart rate was decreased after two weeks transcutaneous vagal nerve stimulation in comparison to sham treatment (-3.7 beats/min, 95%CI (-6.7:-0.6), P = 0.02).

Conclusion: In this sham-controlled crossover study, we found no evidence that two weeks transcutaneous vagal nerve stimulation induces pain relief in patients with chronic pancreatitis.

Trial registration number: The study is registered at NCT03357029; www.clinicaltrials.gov.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT diagram.
Fig 1. CONSORT diagram.
Study enrolment and randomisation.
Fig 2. Study design & treatment procedure:…
Fig 2. Study design & treatment procedure: Patients were instructed to self-administer the treatment bilaterally on the neck three times per day for two weeks at home.
The treatment was delivered via two stainless steel contact surfaces that were covered with conductive gel before each treatment.
Fig 3
Fig 3
Average pain score during the two study periods for the two sequences (nVNS/sham in black and sham/nVNS in grey). VAS = visual analogue scale.

References

    1. Singh VK, Yadav D, Garg PK. Diagnosis and Management of Chronic Pancreatitis: A Review. JAMA. 2019;322: 2422–2434. 10.1001/jama.2019.19411
    1. Kleeff J, Whitcomb DC, Shimosegawa T, Esposito I, Lerch MM, Gress T, et al.. Chronic pancreatitis. Nat Rev Dis Prim. 2017;3: 1–18. 10.1038/nrdp.2017.60
    1. Chakravarthy K, Chaudhry H, Williams K, Christo PJ. Review of the Uses of Vagal Nerve Stimulation in Chronic Pain Management. Current Pain and Headache Reports. 2015. 10.1007/s11916-015-0528-6
    1. Garcia RG, Lin RL, Lee J, Kim J, Barbieri R, Sclocco R, et al.. Modulation of brainstem activity and connectivity by respiratory-gated auricular vagal afferent nerve stimulation in migraine patients. Pain. 2017;158: 1461–1472. 10.1097/j.pain.0000000000000930
    1. Napadow V, Edwards RR, Cahalan CM, Mensing G, Greenbaum S, Valovska A, et al.. Evoked Pain Analgesia in Chronic Pelvic Pain Patients Using Respiratory-Gated Auricular Vagal Afferent Nerve Stimulation. Pain Med (United States). 2012;13: 777–789. 10.1111/j.1526-4637.2012.01385.x
    1. Juel J, Brock C, Olesen SS, Madzak A, Farmer AD, Aziz Q, et al.. Acute physiological and electrical accentuation of vagal tone has no effect on pain or gastrointestinal motility in chronic pancreatitis. J Pain Res. 2017;10: 1347–1355. 10.2147/JPR.S133438
    1. Botha C, Farmer AD, Nilsson M, Brock C, Gavrila AD, Drewes AM, et al.. Preliminary report: modulation of parasympathetic nervous system tone influences oesophageal pain hypersensitivity. Gut. 2015;64: 611–7. 10.1136/gutjnl-2013-306698
    1. Mwamburi M, Tenaglia AT, Leibler EJ, Staats PS. Review of evidence on noninvasive vagus nerve stimulation for treatment of migraine: efficacy, safety, and implications. Am J Manag Care. 2018;24: S507–S516.
    1. Busch V, Zeman F, Heckel A, Menne F, Ellrich J, Eichhammer P. The effect of transcutaneous vagus nerve stimulation on pain perception-An experimental study. Brain Stimul. 2013;6: 202–209. 10.1016/j.brs.2012.04.006
    1. Muthulingam JA, Olesen SS, Hansen TM, Brock C, Drewes AM, Frøkjær JB. Study protocol for a randomised double-blinded, sham-controlled, prospective, cross-over clinical trial of vagal neuromodulation for pain treatment in patients with chronic pancreatitis. BMJ Open. 2019;9: e029546. 10.1136/bmjopen-2019-029546
    1. Layer P, Yamamoto H, Kalthoff L, Clain J, Bakken L, DiMagno E. The different courses of early- and late-onset idiopathic and alcoholic chronic pancreatitis. Gastroenterology. 1994;107: 1481–7. 10.1016/0016-5085(94)90553-3
    1. Tassorelli C, Grazzi L, Tommaso M De, Pierangeli G. Noninvasive vagus nerve stimulation as acute therapy for migraine The randomized PRESTO study. 2018;0. 10.1212/WNL.0000000000005857
    1. Use I, Risks P. Instructions for Use for gammaCore ® -S 1: 1–28.
    1. Jensen MP, Turner JA, Romano JM, Fisher LD. Comparative reliability and validity of chronic pain intensity measures. Pain. 1999;83: 157–162. 10.1016/s0304-3959(99)00101-3
    1. Aaronson NK, Ahmedzai S, Bergman B, et al.. The European organization for research and treatment of cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. Journal of the National Cancer Institute. 1993. pp. 365–376. 10.1093/jnci/85.5.365
    1. Cleeland CS. Brief Pain Inventory (BPI). Cleel CS MD Anderson Cancer Cent. 1982;1100: 6. 10.1007/978-1-4419-9893-4_13
    1. Farrar JT, Young JP, LaMoreaux L, Werth JL, Poole RM. Clinical importance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale. Pain. 2001;94: 149–158. 10.1016/S0304-3959(01)00349-9
    1. Farmer AD, Coen SJ, Kano M, Weltens N, Ly HG, Botha C, et al.. Normal values and reproducibility of the real-time index of vagal tone in healthy humans: a multi-center study. Ann Gastroenterol. 2014;27: 362–368.
    1. Gilron I, Bailey JM, Tu D, Holden RR, Weaver DF, Houlden RL. Morphine, Gabapentin, or Their Combination for Neuropathic Pain. N Engl J Med. 2005;352: 1324–1334. 10.1056/NEJMoa042580
    1. Wellek S, Blettner M. Vom richtigen Umgang mit dem Crossover-Design in klinischen Studien: Teil 18 der Serie zur Bewertung wissenschaftlicher Publikationen. Dtsch Arztebl Int. 2012;109: 276–281. 10.3238/arztebl.2012.0276
    1. Olesen SS, Bouwense S a W, Wilder-Smith OHG, van Goor H, Drewes AM. Pregabalin reduces pain in patients with chronic pancreatitis in a randomized, controlled trial. Gastroenterology. 2011;141: 536–43. 10.1053/j.gastro.2011.04.003
    1. Pham A, Forsmark C. Chronic pancreatitis: review and update of etiology, risk factors, and management. F1000Research. 2018;7: 607. 10.12688/f1000research.12852.1
    1. Drewes AM, Kempeneers MA, Andersen DK, Arendt-Nielsen L, Besselink MG, Boermeester MA, et al.. Controversies on the endoscopic and surgical management of pain in patients with chronic pancreatitis: pros and cons! Gut. 2019;68: 1343–1351. 10.1136/gutjnl-2019-318742
    1. Lange G, Janal MN, Maniker A, Fitzgibbons J, Fobler M, Cook D, et al.. Safety and efficacy of vagus nerve stimulation in fibromyalgia: a phase I/II proof of concept trial. Pain Med. 2011;12: 1406–1413. 10.1111/j.1526-4637.2011.01203.x
    1. Gottfried-Blackmore A, Adler EP, Fernandez-Becker N, Clarke J, Habtezion A, Nguyen L. Open-label pilot study: Non-invasive vagal nerve stimulation improves symptoms and gastric emptying in patients with idiopathic gastroparesis. Neurogastroenterol Motil. 2019;32: e13769. 10.1111/nmo.13769
    1. Paulon E, Nastou D, Jaboli F, Marin J, Liebler E, Epstein O. Proof of concept: short-term non-invasive cervical vagus nerve stimulation in patients with drug-refractory gastroparesis. Frontline Gastroenterol. 2017;8: 325–330. 10.1136/flgastro-2017-100809
    1. Barbanti P, Grazzi L, Egeo G, Padovan AM, Liebler E, Bussone G. Non-invasive vagus nerve stimulation for acute treatment of high-frequency and chronic migraine: an open-label study. J Headache Pain. 2015;16. 10.1186/s10194-015-0542-4
    1. Oshinsky ML, Murphy AL, Hekierski HJ, Cooper M, Simon BJ. Noninvasive vagus nerve stimulation as treatment for trigeminal allodynia. Pain. 2014;155: 1037–1042. 10.1016/j.pain.2014.02.009
    1. Frangos E, Komisaruk BR. Access to Vagal Projections via Cutaneous Electrical Stimulation of the Neck: fMRI Evidence in Healthy Humans. Brain Stimul. 2017;10: 19–27. 10.1016/j.brs.2016.10.008
    1. Brock C, Brock B, Aziz Q, Møller HJ, Pfeiffer Jensen M, Drewes AM, et al.. Transcutaneous cervical vagal nerve stimulation modulates cardiac vagal tone and tumor necrosis factor-alpha. Neurogastroenterol Motil. 2017;29: 4–7. 10.1111/nmo.12999
    1. Liu H, Yang Z, Huang L, Qu W, Hao H, Li L. Heart-rate variability indices as predictors of the response to vagus nerve stimulation in patients with drug-resistant epilepsy. Epilepsia. 2017;58: 1015–1022. 10.1111/epi.13738
    1. Matteoli G, Boeckxstaens GE. The vagal innervation of the gut and immune homeostasis. Gut. 2013;62: 1214–22. 10.1136/gutjnl-2012-302550
    1. Silberstein SD, Calhoun AH, Lipton RB, Grosberg BM, Cady RK, Dorlas S, et al.. Chronic migraine headache prevention with noninvasive vagus nerve stimulation: The EVENT study. Neurology. 2016;87: 529–538. 10.1212/WNL.0000000000002918
    1. Sator-Katzenschlager SM, Scharbert G, Kozek-Langenecker SA, Szeles JC, Finster G, Schiesser AW, et al.. The Short- and Long-Term Benefit in Chronic Low Back Pain Through Adjuvant Electrical Versus Manual Auricular Acupuncture. Anesth Analg. 2004. 10.1213/01.ane.0000107941.16173.f7
    1. Grazzi L, Tassorelli C, De Tommaso M, Pierangeli G, Martelletti P, Rainero I, et al.. Practical and clinical utility of non-invasive vagus nerve stimulation (nVNS) for the acute treatment of migraine: A post hoc analysis of the randomized, sham-controlled, double-blind PRESTO trial. J Headache Pain. 2018;19: 1–9. 10.1186/s10194-017-0831-1
    1. Piao WH, Campagnolo D, Dayao C, Lukas RJ, Wu J, Shi FD. Nicotine and inflammatory neurological disorders. Acta Pharmacol Sin. 2009;30: 715–722. 10.1038/aps.2009.67

Source: PubMed

3
S'abonner