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

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

Introduction: The management of chronic pancreatitis (CP) is challenging and requires a personalised approach focused on the individual patient's main symptoms. Abdominal pain is the most prominent symptom in CP, where central pain mechanisms, including sensitisation and impaired pain modulation, often are involved. Recent clinical studies suggest that vagal nerve stimulation (VNS) induces analgesic effects through the modulation of central pain pathways. This study aims to investigate the effect of 2 weeks transcutaneous VNS (t-VNS) on clinical pain in patients with CP, in comparison to the effect of sham treatment.

Methods and analysis: Twenty-one patients with CP will be enrolled in this randomised, double-blinded, single-centre, sham-controlled, cross-over study. The study has two treatment periods: A 2-week active t-VNS using GammaCore device and a 2-week treatment with a sham device. During both treatment periods, the patients are instructed to self-administer VNS bilaterally to the cervical vagal area, three times per day. Treatment periods will be separated by 2 weeks. During the study period, patients will record their daily pain experience in a diary (primary clinical endpoint). In addition, all subjects will undergo testing which will include MRI, quantitative sensory testing, cardiac vagal tone assessment and collecting blood samples, before and after the two treatments to investigate mechanisms underlying VNS effects. The data will be analysed using the principle of intention to treat.

Ethics and dissemination: The regional ethics committee has approved the study: N-20170023. Results of the trial will be submitted for publication in peer-reviewed journals.

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

Keywords: chronic pain; clinical trials; gastrointestinal disease; transcutaneous electrical nerve stimulation; viscera.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Mode of action of transcutaneous vagal nerve stimulation (t-VNS). (1) Pain arises in the periphery, for example, pancreas and a signal is sent to the spinal cord. This leads to the ascending activation of the spinal neurons (2). In the brain, pain is processed in higher cortical centres (3). t-VNS, it is expected to block the perception of pain in the cerebral cortex, by stimulating nucleus tractus solitarius and thereby decrease glutamate level. Simultaneously, the net-descending inhibition will be activated as a result of top-down input from cortex and the limbic system (4).
Figure 2
Figure 2
Schematic flow chart of the interventional study design enabling comparison of the modulatory effect to self-administered of transcutaneous vagal nerve stimulation (t-VNS) in patients with chronic pancreatitis. Patients with chronic pancreatitis will be randomly assigned to one of two double-blinded treatments: (1) 2 weeks of t-VNS, 2 weeks of washout and 2 weeks of sham treatment; or (2) 2 weeks of sham treatment, 2 weeks of washout and 2 weeks of t-VNS. Evaluation of the two treatments will be assessed by collecting pain diary, pain questionnaires, MRI scan, blood sample, cardiac vagal tone (CVT) and pain assessments. quantitative sensory testing (QST).
Figure 3
Figure 3
Standard Protocol Items: Recommendations for Interventional Trials figure. BPI-SF, Brief Pain Inventory—Short Form; CVT, cardiac vagal tone; PGIC, Patient Global Impression of Changes Questionnaire; QST, quantitative sensory testing; VNS, vagal nerve stimulation.

References

    1. Muniraj T, Aslanian HR, Farrell J, et al. . Chronic pancreatitis, a comprehensive review and update. Part I: epidemiology, etiology, risk factors, genetics, pathophysiology, and clinical features. Dis Mon 2014;60:530–50. 10.1016/j.disamonth.2014.11.002
    1. Pham A, Forsmark C. Chronic pancreatitis: review and update of etiology, risk factors, and management. F1000Res 2018;7:607 10.12688/f1000research.12852.1
    1. Mullady DK, Yadav D, Amann ST, et al. . Type of pain, pain-associated complications, quality of life, disability and resource utilisation in chronic pancreatitis: a prospective cohort study. Gut 2011;60:77–84. 10.1136/gut.2010.213835
    1. Botha C, Farmer AD, Nilsson M, 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. Frøkjaer JB, Bergmann S, Brock C, et al. . Modulation of vagal tone enhances gastroduodenal motility and reduces somatic pain sensitivity. Neurogastroenterol Motil 2016;28:592–8. 10.1111/nmo.12760
    1. Johnson RL, Wilson CG. A review of vagus nerve stimulation as a therapeutic intervention. J Inflamm Res 2018;11:203–13. 10.2147/JIR.S163248
    1. Napadow V, Edwards RR, Cahalan CM, et al. . Evoked pain analgesia in chronic pelvic pain patients using respiratory-gated auricular vagal afferent nerve stimulation. Pain Med 2012;13:777–89. 10.1111/j.1526-4637.2012.01385.x
    1. Brock C, Brock B, Aziz Q, et al. . Transcutaneous cervical vagal nerve stimulation modulates cardiac vagal tone and tumor necrosis factor-alpha. Neurogastroenterol Motil 2017;29:e12999–7. 10.1111/nmo.12999
    1. Chakravarthy K, Chaudhry H, Williams K, et al. . Review of the uses of vagal nerve stimulation in chronic pain management. Curr Pain Headache Rep 2015;19:54 10.1007/s11916-015-0528-6
    1. Frangos E, Ellrich J, Komisaruk BR. Non-invasive access to the vagus nerve central projections via electrical stimulation of the external ear: fMRI evidence in humans. Brain Stimul 2015;8:624–36. 10.1016/j.brs.2014.11.018
    1. Yuan H, Silberstein SD. Vagus Nerve and Vagus Nerve Stimulation, a Comprehensive Review: Part II. Headache 2016;56:259–66. 10.1111/head.12650
    1. Gaul C, Diener HC, Silver N, et al. . Non-invasive vagus nerve stimulation for PREVention and acute treatment of chronic cluster headache (PREVA): a randomised controlled study. Cephalalgia 2016;36:534–46. 10.1177/0333102415607070
    1. Moher D, Hopewell S, Schulz KF, et al. . CONSORT 2010 explanation and elaboration: updated guidelines for reporting parallel group randomised trials. Int J Surg 2012;10:28–55. 10.1016/j.ijsu.2011.10.001
    1. Chan AW, Tetzlaff JM, Gøtzsche PC, et al. . SPIRIT 2013 explanation and elaboration: guidance for protocols of clinical trials. BMJ 2013;346:e7586 10.1136/bmj.e7586
    1. Vitória A, De AL, Ribeiro V, et al. . Effects of high-frequency transcranial magnetic stimulation on functional performance in individuals with incomplete spinal cord injury : study protocol for a randomized controlled trial. 2017:1–11.
    1. Kuppuswamy A, Balasubramaniam AV, Maksimovic R, et al. . Action of 5 Hz repetitive transcranial magnetic stimulation on sensory, motor and autonomic function in human spinal cord injury. Clin Neurophysiol 2011;122:2452–61. 10.1016/j.clinph.2011.04.022
    1. Layer P, Yamamoto H, Kalthoff L, et al. . 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. Strickland I, Mwamburi M, Davis S, et al. . Noninvasive vagus nerve stimulation in a primary care setting: effects on quality of life and utilization measures in multimorbidity patients with or without primary headache. Am J Manag Care 2018;24(24 Suppl):S517–S526.
    1. Grazzi L, Tassorelli C, de Tommaso M, 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/s10194-018-0928-1
    1. Tarn J, Legg S, Mitchell S, et al. . The effects of noninvasive vagus nerve stimulation on fatigue and immune responses in patients with primary Sjögren’s Syndrome. Neuromodulation 2018;2018.
    1. Nesbitt AD, Marin JC, Tompkins E, et al. . Initial use of a novel noninvasive vagus nerve stimulator for cluster headache treatment. Neurology 2015;84:1249–53. 10.1212/WNL.0000000000001394
    1. De Icco R, Martinelli D, Bitetto V, et al. . Peripheral vagal nerve stimulation modulates the nociceptive withdrawal reflex in healthy subjects: A randomized, cross-over, sham-controlled study. Cephalalgia 2018;38:1658–64. 10.1177/0333102417742347
    1. Silberstein SD, Mechtler LL, Kudrow DB, et al. . Non-Invasive Vagus Nerve Stimulation for the ACute Treatment of Cluster Headache: Findings From the Randomized, Double-Blind, Sham-Controlled ACT1 Study. Headache 2016;56:1317–32. 10.1111/head.12896
    1. Use I, Risks P. Instructions for Use for gammaCore ® -S 1: 1-28.
    1. Fayers PM. EORTC QLQ-C30 scoring manual the EORTC QLQ-C30 introduction. EORTC QLQ-C30 Scoring Man EORTC QLQ-C30 Introd 2001;30:1–67.
    1. Cleeland CS. Brief Pain Inventory (BPI). Cleel CS MD Anderson Cancer Cent 1982;1100:6.
    1. Farrar JT, Young JP, LaMoreaux L, et al. . Clinical importance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale. Pain 2001;94:149–58. 10.1016/S0304-3959(01)00349-9
    1. Arendt-Nielsen L, Morlion B, Perrot S, et al. . Assessment and manifestation of central sensitisation across different chronic pain conditions. Eur J Pain 2018;22:216–41. 10.1002/ejp.1140
    1. Olesen SS, Bouwense SA, Wilder-Smith OH, et al. . 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. Kennedy DL, Kemp HI, Ridout D, et al. . Reliability of conditioned pain modulation: a systematic review. Pain 2016;157:2410–9. 10.1097/j.pain.0000000000000689
    1. Yarnitsky D, Bouhassira D, Drewes AM, et al. . Recommendations on practice of conditioned pain modulation (CPM) testing. Eur J Pain 2015;19:805–6. 10.1002/ejp.605
    1. Farmer AD, Coen SJ, Kano M, et al. . Normal values and reproducibility of the real-time index of vagal tone in healthy humans: a multi-center study. Ann Gastroenterol 2014;27:362–8.
    1. Trial C, Olesen SS, Bouwense SAW, et al. . Clinical — pancreas pregabalin reduces pain in patients with chronic pancreatitis in a. YGAST 2011;141:536–43.
    1. College Station TSL. StataCorp. 2015: Stata Stat Softw Release, 2013.
    1. Harris PA, Taylor R, Thielke R, et al. . Research electronic data capture (REDCap)--a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform 2009;42:377–81. 10.1016/j.jbi.2008.08.010
    1. Hansen TM, Olesen AE, Simonsen CW, et al. . Cingulate metabolites during pain and morphine treatment as assessed by magnetic resonance spectroscopy. J Pain Res 2014;7:269–76. 10.2147/JPR.S61193
    1. Dimcevski G, Sami SA, Funch-Jensen P, et al. . Pain in chronic pancreatitis: the role of reorganization in the central nervous system. Gastroenterology 2007;132:1546–56. 10.1053/j.gastro.2007.01.037
    1. Frøkjær JB, Olesen SS, Gram M, et al. . Altered brain microstructure assessed by diffusion tensor imaging in patients with chronic pancreatitis. Gut 2011;60:1554–62. 10.1136/gut.2010.236620
    1. Frøkjær JB, Bouwense SA, Olesen SS, et al. . Reduced cortical thickness of brain areas involved in pain processing in patients with chronic pancreatitis. Clin Gastroenterol Hepatol 2012;10:434–8. 10.1016/j.cgh.2011.11.024
    1. Lelic D, Olesen SS, Graversen C, et al. . Electrophysiology as a tool to unravel the origin of pancreatic pain. World J Gastrointest Pathophysiol 2014;5:33–9. 10.4291/wjgp.v5.i1.33
    1. Muthulingam J, Olesen SS, Hansen TM, et al. . Progression of Structural Brain Changes in Patients With Chronic Pancreatitis and Its Association to Chronic Pain: A 7-Year Longitudinal Follow-up Study. Pancreas 2018;47:1267–76. 10.1097/MPA.0000000000001151
    1. Apkarian AV, Hashmi JA, Baliki MN. Pain and the brain: specificity and plasticity of the brain in clinical chronic pain. Pain 2011;152(3 Suppl):S49–64. 10.1016/j.pain.2010.11.010
    1. Garcia RG, Lin RL, Lee J, et al. . Modulation of brainstem activity and connectivity by respiratory-gated auricular vagal afferent nerve stimulation in migraine patients. Pain 2017;158:1461–72. 10.1097/j.pain.0000000000000930
    1. O’Connell NE, Marston L, Spencer S, et al. . Non-invasive brain stimulation techniques for chronic pain. Cochrane Database Syst Rev 2018;3:CD008208 10.1002/14651858.CD008208.pub4
    1. Seminowicz DA, Wideman TH, Naso L, et al. . Effective treatment of chronic low back pain in humans reverses abnormal brain anatomy and function. J Neurosci 2011;31:7540–50. 10.1523/JNEUROSCI.5280-10.2011
    1. Čeko M, Shir Y, Ouellet JA, et al. . Partial recovery of abnormal insula and dorsolateral prefrontal connectivity to cognitive networks in chronic low back pain after treatment. Hum Brain Mapp 2015;36:2075–92. 10.1002/hbm.22757
    1. Bonaz B, Sinniger V, Pellissier S. Vagal tone: effects on sensitivity, motility, and inflammation. Neurogastroenterol Motil 2016;28:455–62. 10.1111/nmo.12817
    1. Watkins LR, Maier SF, Goehler LE. Cytokine-to-brain communication: a review & analysis of alternative mechanisms. Life Sci 1995;57:1011–26. 10.1016/0024-3205(95)02047-M
    1. Hosoi T, Okuma Y, Matsuda T, et al. . Novel pathway for LPS-induced afferent vagus nerve activation: possible role of nodose ganglion. Auton Neurosci 2005;120:104–7. 10.1016/j.autneu.2004.11.012

Source: PubMed

3
S'abonner