Remote Electrical Neuromodulation (REN) Relieves Acute Migraine: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial

David Yarnitsky, David W Dodick, Brian M Grosberg, Rami Burstein, Alon Ironi, Dagan Harris, Tamar Lin, Stephen D Silberstein, David Yarnitsky, David W Dodick, Brian M Grosberg, Rami Burstein, Alon Ironi, Dagan Harris, Tamar Lin, Stephen D Silberstein

Abstract

Objective: To assess the efficacy and safety of a remote electrical neuromodulation (REN) device for the acute treatment of migraine.

Background: There is a significant unmet need for novel effective well-tolerated acute migraine treatments. REN is a novel acute migraine treatment that stimulates upper arm peripheral nerves to induce conditioned pain modulation - an endogenous analgesic mechanism in which conditioning stimulation inhibits pain in remote body regions. A recent pilot study showed that REN can significantly reduce headache. We have conducted a randomized, double-blind, sham-controlled study to further evaluate the efficacy and safety of REN for the acute treatment of migraine.

Methods: This was a randomized, double-blind, sham-controlled, multicenter study conducted at 7 sites in the United States and 5 sites in Israel. Two hundred and fifty-two adults meeting the International Classification of Headache Disorders criteria for migraine with 2-8 migraine headaches per month were randomized in a 1:1 ratio to active or sham stimulation. A smartphone-controlled wireless device was applied for 30-45 minutes on the upper arm within 1 hour of attack onset; electrical stimulation was at a perceptible but non-painful intensity level. Migraine pain levels were recorded at baseline, 2, and 48 hours post-treatment. Most bothersome symptoms (MBS) were also recorded. The primary efficacy endpoint was the proportion of participants achieving pain relief at 2 hours post-treatment (improvement from severe or moderate pain to mild or none, or from mild pain to none). Relief of MBS and pain-free at 2 hours were key secondary endpoints.

Results: Active stimulation was more effective than sham stimulation in achieving pain relief (66.7% [66/99] vs 38.8% [40/103]; therapeutic gain of 27.9% [CI95% , 15.6-40.2]; P < .0001), pain-free (37.4% vs 18.4%, P = .003), and MBS relief (46.3% vs 22.2%, P = .0008) at 2 hours post-treatment. The pain relief and pain-free superiority of the active treatment was sustained 48 hours post-treatment. The incidence of device-related adverse events was low and similar between treatment groups (4.8% [6/126] vs 2.4% [3/126], P = .499).

Conclusions: REN provides superior clinically meaningful relief of migraine pain and MBS compared to placebo, offering a safe and effective non-pharmacological alternative for acute migraine treatment.

Trial registration: ClinicalTrials.gov NCT03361423.

Keywords: conditioned pain modulation; headache; migraine; neuromodulation; non-pharmacological treatment; remote electrical neuromodulation.

© 2019 The Authors. Headache: The Journal of Head and Face Pain published by Wiley Periodicals, Inc. on behalf of American Headache Society.

Figures

Figure 1
Figure 1
Schematic illustration of the principle of operation of REN. The device stimulates C and Aδ noxious sensory fibers of the upper arm above their depolarization thresholds but below the perceived pain threshold. The noxious information reaches the brainstem through the ascending pain pathway (black). This information activates the descending pain inhibitory pathway (green), involving the brainstem pain regulation center (which includes the PAG, RVM, and subnucleus reticularis dorsalis [SRD]), and the release of serotonin and noradrenalin, which inhibit incoming messages of pain in the trigeminal cervical complex (TCC) that occur during a headache of a migraine attack (red). PAG = periaqueductal gray; RVM = rostral ventromedial medulla; SRD = subnucleus reticularis dorsalis; TCC = trigeminal cervical complex.
Figure 2
Figure 2
Enrollment and randomization of participants. mITT = modified intention to treat.
Figure 3
Figure 3
Efficacy endpoints. (A) Pain response at 2 and 48 hours post‐treatment. (B) MBS response at 2 hours post‐treatment. The error bars represent 95% confidence intervals. ***P < .001, **P < .005, *P < .05. MBS = most bothersome symptom.

References

    1. Vos T, Abajobir AA, Abate KH, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990‐2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017;390:1211‐1259.
    1. Society (IHS) HCC of the IH . The international classification of headache disorders, (beta version). Cephalalgia. 2013;33:629‐808.
    1. Silva AND, Tepper SJ. Acute treatment of migraines. CNS Drugs. 2012;26:823‐839.
    1. Goadsby PJ, Sprenger T. Current practice and future directions in the prevention and acute management of migraine. Lancet Neurol. 2010;9:285‐298.
    1. Diener H‐C, Limmroth V. Medication‐overuse headache: A worldwide problem. Lancet Neurol. 2004;3:475‐483.
    1. Bigal ME, Lipton RB. Overuse of acute migraine medications and migraine chronification. Curr Pain Headache Rep. 2009;13:301‐307.
    1. Katić BJ, Krause SJ, Tepper SJ, Hu HX, Bigal ME. Adherence to acute migraine medication: What does it mean, why does it matter? Headache. 2010;50:117‐129.
    1. Bigal M, Krymchantowski AV, Lipton RB. Barriers to satisfactory migraine outcomes. What have we learned, where do we stand? Headache. 2009;49:1028‐1041.
    1. Alam A, Munjal S, Reed M, et al. Triptan use and discontinuation in a representative sample of persons with migraine: Results from migraine in America symptoms and treatment (MAST) study (P4.10-019). Neurology. 2019;92(Suppl. 15) P4.10-019.
    1. Puledda F, Goadsby PJ. An update on non‐pharmacological neuromodulation for the acute and preventive treatment of migraine. Headache. 2017;57:685‐691.
    1. Starling A. Noninvasive neuromodulation in migraine and cluster headache. Curr Opin Neurol. 2018;31:268‐273.
    1. Nir R‐R, Yarnitsky D. Conditioned pain modulation. Curr Opin Support Palliat Care. 2015;9:131‐137.
    1. Yarnitsky D, Volokh L, Ironi A, et al. Nonpainful remote electrical stimulation alleviates episodic migraine pain. Neurology. 2017;88:1250‐1255.
    1. Demiryurek BE, Ertem DH, Tekin A, Ceylan M, Aras YG, Gungen BD. Effects of onabotulinumtoxinA treatment on efficacy, depression, anxiety, and disability in Turkish patients with chronic migraine. Neurol Sci. 2016;37:1779‐1784.
    1. Benjamini Y, Krieger AM, Yekutieli D. Adaptive linear step‐up procedures that control the false discovery rate. Biometrika. 2006;93:491‐507.
    1. Tassorelli C, Grazzi L, De Tommaso M, et al. Noninvasive vagus nerve stimulation as acute therapy for migraine: The randomized PRESTO study. Neurology. 2018;91:e364‐e373.
    1. Spierings EL, Brandes JL, Kudrow DB, et al. Randomized, double‐blind, placebo‐controlled, parallel‐group, multi‐center study of the safety and efficacy of ADAM zolmitriptan for the acute treatment of migraine. Cephalalgia. 2018;38:215‐224.
    1. Diener H‐C, Barbanti P, Dahlöf C, et al. BI 44370 TA, an oral CGRP antagonist for the treatment of acute migraine attacks: Results from a phase II study. Cephalalgia. 2010;31:573‐584.
    1. Ossipov MH, Dussor GO, Porreca F. Central modulation of pain. J Clin Invest. 2010;120:3779‐3787.
    1. Schuh‐Hofer S, Fischer J, Unterberg A, Treede RD, Ahmadi R. Spinal cord stimulation modulates descending pain inhibition and temporal summation of pricking pain in patients with neuropathic pain. Acta Neurochir. 2018;160:2509‐2519.
    1. Yarnitsky D, Arendt‐Nielsen L, Bouhassira D, et al. Recommendations on terminology and practice of psychophysical DNIC testing. Eur J Pain. 2010;14:339‐339.
    1. Le Bars D, Dickenson AH, Besson J‐M. Diffuse noxious inhibitory controls (DNIC). I. Effects on dorsal horn convergent neurones in the rat. Pain. 1979;6:283‐304.
    1. Lautenbacher S, Roscher S, Strian F. Inhibitory effects do not depend on the subjective experience of pain during heterotopic noxious conditioning stimulation (HNCS): A contribution to the psychophysics of pain inhibition. Eur J Pain. 2002;6:365‐374.
    1. Melzack R, Wall PD. Pain mechanisms: A new theory. Science. 1965;150:971‐979.
    1. Lipton RB, Dodick DW, Silberstein SD, et al. Single‐pulse transcranial magnetic stimulation for acute treatment of migraine with aura: A randomised, double‐blind, parallel‐group, sham‐controlled trial. Lancet Neurol. 2010;9:373‐380.
    1. Chou DE, Shnayderman Yugrakh M, Winegarner D, Rowe V, Kuruvilla D, Schoenen J. Acute migraine therapy with external trigeminal neurostimulation (ACME): A randomized controlled trial. Cephalalgia. 2019;39:3‐14.
    1. Cameron C, Kelly S, Hsieh S‐C, et al. Triptans in the acute treatment of migraine: A systematic review and network meta‐analysis. Headache. 2015;55:221‐235.
    1. Holland PR, Goadsby PJ. Targeted CGRP small molecule antagonists for acute migraine therapy. Neurotherapeutics. 2018;15:304‐312.
    1. Wietecha LA, Kuca B, Asafu‐Adjei J. Phase 3 studies (SAMURAI, SPARTAN) of lasmiditan compared to placebo for acute treatment of migraine (S50. 008). Neurology. 2018;90(Suppl. 15):S50‐008.
    1. Ferrari MD, Roon KI, Lipton RB, Goadsby PJ. Oral triptans (serotonin 5‐HT1B/1D agonists) in acute migraine treatment: A meta‐analysis of 53 trials. Lancet. 2001;358:1668‐1675.
    1. Silberstein SD, Calhoun AH, Lipton RB, et al. Chronic migraine headache prevention with noninvasive vagus nerve stimulation: The EVENT study. Neurology. 2016;87:529‐538.
    1. Lipton RB, Pearlman SH. Transcranial magnetic simulation in the treatment of migraine. Neurotherapeutics. 2010;7:204‐212.
    1. Asano E, Goadsby PJ. How do we fashion better trials for neurostimulator studies in migraine? Neurology. 2013;80:694.

Source: PubMed

3
Abonneren