425
Views
4
CrossRef citations to date
0
Altmetric
Clinical Trial Report

Low-Dose Dextromethorphan for the Treatment of Fibromyalgia Pain: Results from a Longitudinal, Single-Blind, Placebo-Controlled Pilot Trial

, &
Pages 189-200 | Published online: 27 Jan 2021

References

  • Wolfe F, Clauw DJ, Fitzcharles MA, et al. 2016 Revisions to the 2010/2011 fibromyalgia diagnostic criteria. Semin Arthritis Rheum. 2016;46(3):319–329. doi:10.1016/j.semarthrit.2016.08.012
  • Wolfe F, Clauw DJ, Fitzcharles M-A, et al. The American College of Rheumatology preliminary diagnostic criteria for fibromyalgia and measurement of symptom severity. Arthritis Care Res (Hoboken). 2010;62(5):600–610. doi:10.1002/acr.20140
  • Staud R, Cannon RC, Mauderli AP, Robinson ME, Price DD, Vierck CJ. Temporal summation of pain from mechanical stimulation of muscle tissue in normal controls and subjects with fibromyalgia syndrome. Pain. 2003;102(1–2):87–95. doi:10.1016/s0304-3959(02)00344-5
  • Staud R, Craggs JG, Perlstein WM, Robinson ME, Price DD. Brain activity associated with slow temporal summation of C-fiber evoked pain in fibromyalgia patients and healthy controls. Eur J Pain. 2008;12(8):1078–1089. doi:10.1016/j.ejpain.2008.02.002
  • Staud R, Robinson ME, Price DD. Temporal summation of second pain and its maintenance are useful for characterizing widespread central sensitization of fibromyalgia patients. J Pain. 2007;8(11):893–901. doi:10.1016/j.jpain.2007.06.006
  • Staud R, Vierck CJ, Cannon RL, Mauderli AP, Price DD. Abnormal sensitization and temporal summation of second pain (wind-up) in patients with fibromyalgia syndrome. Pain. 2001;91(1–2):165–175. doi:10.1016/S0304-3959(00)00432-2
  • Staud R, Weyl EE, Riley JL, Fillingim RB. Slow temporal summation of pain for assessment of central pain sensitivity and clinical pain of fibromyalgia patients. PLoS One. 2014;9(2):e89086. doi:10.1371/journal.pone.0089086
  • Cagnie B, Coppieters I, Denecker S, Six J, Danneels L, Meeus M. Central sensitization in fibromyalgia? A systematic review on structural and functional brain MRI. Semin Arthritis Rheum. 2014;44(1):68–75. doi:10.1016/j.semarthrit.2014.01.001
  • Lund I, Lundeberg T, Carleson J, Sönnerfors H, Uhrlin B, Svensson E. Corticotropin releasing factor in urine–a possible biochemical marker of fibromyalgia. Responses to massage and guided relaxation. Neurosci Lett. 2006;403(1–2):166–171. doi:10.1016/j.neulet.2006.04.038
  • McLean SA, Williams DA, Stein PK, et al. Cerebrospinal fluid corticotropin-releasing factor concentration is associated with pain but not fatigue symptoms in patients with fibromyalgia. Neuropsychopharmacology. 2006;31(12):2776–2782. doi:10.1038/sj.npp.1301200
  • Becker S, Schweinhardt P. Dysfunctional neurotransmitter systems in fibromyalgia, their role in central stress circuitry and pharmacological actions on these systems. Pain Res Treat. 2012;2012:741746. doi:10.1155/2012/741746
  • Stahl SM. Fibromyalgia–pathways and neurotransmitters. Hum Psychopharmacol. 2009;24(Suppl 1):S11–17. doi:10.1002/hup.1029
  • Harris RE. Elevated excitatory neurotransmitter levels in the fibromyalgia brain. Arthritis Res Ther. 2010;12(5):141. doi:10.1186/ar3136
  • Russell IJ, Orr MD, Littman B, et al. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum. 1994;37(11):1593–1601. doi:10.1002/art.1780371106
  • Bidari A, Ghavidel-Parsa B, Rajabi S, Sanaei O, Toutounchi M. The acute effect of maximal exercise on plasma beta-endorphin levels in fibromyalgia patients. Korean J Pain. 2016;29(4):249–254. doi:10.3344/kjp.2016.29.4.249
  • Cording M, Derry S, Phillips T, Moore RA, Wiffen PJ. Milnacipran for pain in fibromyalgia in adults. Cochrane Database Syst Rev. 2015;2015(10):Cd008244.
  • Arnold LM, Hess EV, Hudson JI, Welge JA, Berno SE, Keck PE. A randomized, placebo-controlled, double-blind, flexible-dose study of fluoxetine in the treatment of women with fibromyalgia. Am J Med. 2002;112(3):191–197. doi:10.1016/S0002-9343(01)01089-0
  • Walitt B, Urrútia G, Nishishinya MB, Cantrell SE, Häuser W. Selective serotonin reuptake inhibitors for fibromyalgia syndrome. Cochrane Database Syst Rev. 2015;2015(6):Cd011735.
  • Harris RE, Clauw DJ, Scott DJ, McLean SA, Gracely RH, Zubieta JK. Decreased central mu-opioid receptor availability in fibromyalgia. J Neurosci. 2007;27(37):10000–10006. doi:10.1523/JNEUROSCI.2849-07.2007
  • Rao SG, Clauw DJ. The management of fibromyalgia. Drugs Today. 2004;40(6):539–554. doi:10.1358/dot.2004.40.6.850485
  • Goldenberg DL, Clauw DJ, Palmer RE, Clair AG. Opioid use in fibromyalgia: a cautionary tale. Mayo Clin Proc. 2016;91(5):640–648. doi:10.1016/j.mayocp.2016.02.002
  • Fitzcharles M-A, Faregh N, Ste-Marie PA, Shir Y. Opioid use in fibromyalgia is associated with negative health related measures in a prospective cohort study. Pain Res Treat. 2013;2013:898493. doi:10.1155/2013/898493
  • Peng X, Robinson RL, Mease P, et al. Long-term evaluation of opioid treatment in fibromyalgia. Clin J Pain. 2015;31(1):7–13. doi:10.1097/AJP.0000000000000079
  • Albrecht DS, Forsberg A, Sandström A, et al. Brain glial activation in fibromyalgia - a multi-site positron emission tomography investigation. Brain Behav Immun. 2019;75:72–83. doi:10.1016/j.bbi.2018.09.018
  • Davalos D, Grutzendler J, Yang G, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8(6):752–758. doi:10.1038/nn1472
  • Ransohoff RM, Perry VH. Microglial physiology: unique stimuli, specialized responses. Annu Rev Immunol. 2009;27:119–145. doi:10.1146/annurev.immunol.021908.132528
  • D’Ambrosi N, Finocchi P, Apolloni S, et al. The proinflammatory action of microglial P2 receptors is enhanced in SOD1 models for amyotrophic lateral sclerosis. J Immunol. 2009;183(7):4648–4656. doi:10.4049/jimmunol.0901212
  • Lynch MA. The multifaceted profile of activated microglia. Mol Neurobiol. 2009;40(2):139–156.
  • Sharma M, Arbabzada N, Flood PM. Mechanism underlying beta2-AR agonist-mediated phenotypic conversion of LPS-activated microglial cells. J Neuroimmunol. 2019;332:37–48. doi:10.1016/j.jneuroim.2019.03.017
  • Bianchi M, Sacerdote P, Ricciardi-Castagnoli P, Mantegazza P, Panerai AE. Central effects of tumor necrosis factor alpha and interleukin-1 alpha on nociceptive thresholds and spontaneous locomotor activity. Neurosci Lett. 1992;148(1–2):76–80. doi:10.1016/0304-3940(92)90808-K
  • Aaron LA, Burke MM, Buchwald D. Overlapping conditions among patients with chronic fatigue syndrome, fibromyalgia, and temporomandibular disorder. Arch Intern Med. 2000;160(2):221–227. doi:10.1001/archinte.160.2.221
  • Dantzer R. Cytokine-induced sickness behavior: mechanisms and implications. Ann N Y Acad Sci. 2001;933:222–234. doi:10.1111/j.1749-6632.2001.tb05827.x
  • Dantzer R. Cytokine-induced sickness behaviour: a neuroimmune response to activation of innate immunity. Eur J Pharmacol. 2004;500(1–3):399–411. doi:10.1016/j.ejphar.2004.07.040
  • Dantzer R, Bluthe RM, Gheusi G, et al. Molecular basis of sickness behavior. Ann N Y Acad Sci. 1998;856:132–138. doi:10.1111/j.1749-6632.1998.tb08321.x
  • Poon DC, Ho YS, Chiu K, Wong HL, Chang RC. Sickness: from the focus on cytokines, prostaglandins, and complement factors to the perspectives of neurons. Neurosci Biobehav Rev. 2015;57:30–45. doi:10.1016/j.neubiorev.2015.07.015
  • Patten DK, Schultz BG, Berlau DJ. The safety and efficacy of low-dose naltrexone in the management of chronic pain and inflammation in multiple sclerosis, fibromyalgia, Crohn’s disease, and other chronic pain disorders. Pharmacotherapy. 2018;38(3):382–389. doi:10.1002/phar.2086
  • Younger J, Mackey S. Fibromyalgia symptoms are reduced by low-dose naltrexone: a pilot study. Pain Medicine. 2009;10(4):663–672. doi:10.1111/j.1526-4637.2009.00613.x
  • Parkitny L, Younger J. Reduced pro-inflammatory cytokines after eight weeks of low-dose naltrexone for fibromyalgia. Biomedicines. 2017;5:2. doi:10.3390/biomedicines5020016
  • Cheng W, Li Y, Hou X, et al. Determining the neuroprotective effects of dextromethorphan in lipopolysaccharidestimulated BV2 microglia. Mol Med Rep. 2015;11(2):1132–1138. doi:10.3892/mmr.2014.2794
  • Lee J-H, Choi S-H, Shin T-J, et al. Effect of dextromethorphan on human Kv1.3 channel activity: involvement of C-type inactivation. Eur J Pharmacol. 2011;651(1):122–127. doi:10.1016/j.ejphar.2010.10.091
  • Li G, Cui G, Tzeng N-S, et al. Femtomolar concentrations of dextromethorphan protect mesencephalic dopaminergic neurons from inflammatory damage. FASEB J. 2005;19(6):489–496. doi:10.1096/fj.04-2555com
  • Liu Y, Qin L, Li G, et al. Dextromethorphan protects dopaminergic neurons against inflammation-mediated degeneration through inhibition of microglial activation. J Pharmacol Exp Ther. 2003;305(1):212–218. doi:10.1124/jpet.102.043166
  • Song J-H, Yeh JZ. Dextromethorphan inhibition of voltage-gated proton currents in BV2 microglial cells. Neurosci Lett. 2012;516(1):94–98. doi:10.1016/j.neulet.2012.03.065
  • Zhang W, Qin L, Wang T, et al. 3-hydroxymorphinan is neurotrophic to dopaminergic neurons and is also neuroprotective against LPS-induced neurotoxicity. FASEB J. 2005;19(3):395–397. doi:10.1096/fj.04-1586fje
  • Cohen SP, Verdolin MH, Chang AS, Kurihara C, Morlando BJ, Mao J. The intravenous ketamine test predicts subsequent response to an oral dextromethorphan treatment regimen in fibromyalgia patients. J Pain. 2006;7(6):391–398. doi:10.1016/j.jpain.2005.12.010
  • Journey JD, Stern E. Dextromethorphan Toxicity. Treasure Island (FL): StatPearls Publishing; 2019.
  • Chechneva OV, Mayrhofer F, Daugherty DJ, Pleasure DE, Hong JS, Deng W. Low dose dextromethorphan attenuates moderate experimental autoimmune encephalomyelitis by inhibiting NOX2 and reducing peripheral immune cells infiltration in the spinal cord. Neurobiol Dis. 2011;44(1):63–72. doi:10.1016/j.nbd.2011.06.004
  • Busner J, Targum SD. The clinical global impressions scale: applying a research tool in clinical practice. Psychiatry. 2007;4(7):28–37.
  • Cleeland CS, Ryan KM. Pain assessment: global use of the Brief Pain Inventory. Ann Acad Med Singapore. 1994;23(2):129–138.
  • Stern AF. The Hospital anxiety and depression scale. Occup Med (Chic Ill). 2014;64(5):393–394. doi:10.1093/occmed/kqu024
  • Younger J, Gandhi V, Hubbard E, Mackey S. Development of the Stanford Expectations of Treatment Scale (SETS): a tool for measuring patient outcome expectancy in clinical trials. Clin Trials. 2012;9(6):767–776. doi:10.1177/1740774512465064
  • Skinner HA. Drug Use Questionnaire (DAST-20). Addiction Research Foundation; 1982.
  • Barnhart JW, Massad EN. Determination of dextromethorphan in serum by gas chromatography. J Chromatogr. 1979;163(4):390–395. doi:10.1016/S0378-4347(00)81642-4
  • Marier JF, Deschenes JL, Hage A, et al. Enhancing the uptake of dextromethorphan in the CNS of rats by concomitant administration of the P-gp inhibitor verapamil. Life Sci. 2005;77(23):2911–2926. doi:10.1016/j.lfs.2005.04.025
  • Perry PJ, Fredriksen K, Chew S, et al. The effects of dextromethorphan on driving performance and the standardized field sobriety test. J Forensic Sci. 2015;60(5):1258–1262. doi:10.1111/1556-4029.12833
  • Carter LP, Reissig CJ, Johnson MW, Klinedinst MA, Griffiths RR, Mintzer MZ. Acute cognitive effects of high doses of dextromethorphan relative to triazolam in humans. Drug Alcohol Depend. 2013;128(3):206–213. doi:10.1016/j.drugalcdep.2012.08.025
  • Reissig CJ, Carter LP, Johnson MW, Mintzer MZ, Klinedinst MA, Griffiths RR. High doses of dextromethorphan, an NMDA antagonist, produce effects similar to classic hallucinogens. Psychopharmacology. 2012;223(1):1–15. doi:10.1007/s00213-012-2680-6
  • Pope LE, Schoedel KA, Bartlett C, Sellers EM. A study of potential pharmacokinetic and pharmacodynamic interactions between dextromethorphan/quinidine and memantine in healthy volunteers. Clin Drug Investig. 2012;32(8):e1–15. doi:10.1007/BF03261905
  • de la Coba P, Bruehl S, Moreno-Padilla M, Reyes Del Paso GA. Responses to slowly repeated evoked pain stimuli in fibromyalgia patients: evidence of enhanced pain sensitization. Pain Medicine. 2017;18(9):1778–1786. doi:10.1093/pm/pnw361
  • O’Brien AT, Deitos A, Trinanes Pego Y, Fregni F, Carrillo-de-la-Pena MT. Defective endogenous pain modulation in fibromyalgia: a meta-analysis of temporal summation and conditioned pain modulation paradigms. J Pain. 2018;19(8):819–836. doi:10.1016/j.jpain.2018.01.010
  • Price DD, Staud R, Robinson ME, Mauderli AP, Cannon R, Vierck CJ. Enhanced temporal summation of second pain and its central modulation in fibromyalgia patients. Pain. 2002;99(1–2):49–59. doi:10.1016/S0304-3959(02)00053-2
  • Davies SN, Lodge D. Evidence for involvement of N-methylaspartate receptors in ‘wind-up’ of class 2 neurones in the dorsal horn of the rat. Brain Res. 1987;424(2):402–406. doi:10.1016/0006-8993(87)91487-9
  • Dickenson AH, Sullivan AF. Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C fibre stimulation. Neuropharmacology. 1987;26(8):1235–1238. doi:10.1016/0028-3908(87)90275-9
  • Parada CA, Luccarini P, Woda A. Effect of an NMDA receptor antagonist on the wind-up of neurons in the trigeminal oralis subnucleus. Brain Res. 1997;761(2):313–320. doi:10.1016/S0006-8993(97)00355-7
  • Fayed N, Olivan-Blazquez B, Herrera-Mercadal P, et al. Changes in metabolites after treatment with memantine in fibromyalgia. A double-blind randomized controlled trial with magnetic resonance spectroscopy with a 6-month follow-up. CNS Neurosci Ther. 2014;20(11):999–1007. doi:10.1111/cns.12314
  • Graven-Nielsen T, Aspegren Kendall S, Henriksson KG, et al. Ketamine reduces muscle pain, temporal summation, and referred pain in fibromyalgia patients. Pain. 2000;85(3):483–491. doi:10.1016/S0304-3959(99)00308-5
  • Noppers I, Niesters M, Swartjes M, et al. Absence of long-term analgesic effect from a short-term S-ketamine infusion on fibromyalgia pain: a randomized, prospective, double blind, active placebo-controlled trial. Eur J Pain. 2011;15(9):942–949. doi:10.1016/j.ejpain.2011.03.008
  • Olivan-Blazquez B, Herrera-Mercadal P, Puebla-Guedea M, et al. Efficacy of memantine in the treatment of fibromyalgia: a double-blind, randomised, controlled trial with 6-month follow-up. Pain. 2014;155(12):2517–2525. doi:10.1016/j.pain.2014.09.004
  • Sorensen J, Bengtsson A, Backman E, Henriksson KG, Bengtsson M. Pain analysis in patients with fibromyalgia. Effects of intravenous morphine, lidocaine, and ketamine. Scand J Rheumatol. 1995;24(6):360–365. doi:10.3109/03009749509095181