1,832
Views
30
CrossRef citations to date
0
Altmetric
Review

Emerging concepts on the use of ketamine for chronic pain

, &
Pages 135-146 | Received 08 Sep 2019, Accepted 15 Jan 2020, Published online: 28 Jan 2020

References

  • Domino EF. Taming the ketamine tiger. 1965. Anesthesiology. 2010;113:678–684.
  • Kronenberg RH. Ketamine as an analgesic: parenteral, oral, rectal, subcutaneous, transdermal and intranasal administration. J Pain Palliative Care Pharmacother. 2002;16:27–35.
  • Niesters M, Martini C, Dahan A. Ketamine for chronic pain: risks and benefits. Br J Clin Pharmacol. 2014;77:357–367.
  • Maher DP, Chen L, Mao J. Intravenous ketamine infusions for neuropathic pain management: a promising therapy in need of optimization. Anesth Analg. 2017;124:661–674.
  • Sanacora G, Frye MA, McDonald W, et al. A consensus statement on the use of ketamine in the treatment of mood disorders. JAMA Psychiatry. 2017;74:399–405.
  • Caddy C, Amit BH, McCloud TL, et al. Ketamine and other glutamate receptor modulators for depression in adults. Cochrane Database Syst Rev. 2015;(9). Art. No.: CD011612.
  • Mion G, Le Masson J, Granier C, et al. A retrospective study of ketamine administration and the development of acute or post-traumatic stress disorder in 274 war-wounded soldiers. Anaesthesia. 2017;72:1476–1483.
  • Feder A, Parides MK, Murrough JW, et al. Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA Psychiatry. 2014;71:681–688.
  • Cohen SP, Bhatia A, Buvanendran A, et al. Consensus guidelines on the use of intravenous ketamine infusions for chronic pain from the american society of regional anesthesia and pain medicine, the american academy of pain medicine, and the american society of anesthesiologists. Reg Anesth Pain Med. 2018;43:521–546.
  • Schwenk ES, Viscusi ER, Buvanendran A, et al. Consensus guidelines on the use of intravenous ketamine infusions for acute pain management from the american society of regional anesthesia and pain medicine, the american academy of pain medicine, and the american society of anesthesiologists. Reg Anesth Pain Med. 2018;43:456–466.
  • Thielking M Is the ketamine boom getting out of hand? Scientific American, 2018 Sep 26. 2019, https://www.scientificamerican.com/article/is-the-ketamine-boom-getting-out-of-hand/.
  • Orhurhu V, Orhurhu MS, Bhatia A, et al. Ketamine infusions for chronic pain: a systematic review and meta-analysis of randomized controlled trials. Anesth Analg. 2019 Jul;129(1):241-254.
  • Newport DJ, Carpenter LL, McDonald WM, et al. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172:950–966.
  • Schoevers RA, Chaves TV, Balukova SM, et al. Oral ketamine for the treatment of pain and treatment-resistant depression. Br J Psychiatry. 2016;208:108–113.
  • Popova V, Daly EJ, Trivedi M, et al. Efficacy and safety of flexibly dosed esketamine nasal spray combined with a newly initiated oral antidepressant in treatment-resistant depression: a randomized double-blind active-controlled study. Am J Psychiatry. 2019;176(6):428–438.
  • Brutcher RE, Kurihara C, Bicket MC, et al. Compounded topical pain creams to treat localized chronic pain: a randomized controlled trial. Ann Intern Med. 2019;170(5):309–318.
  • Finnerup NB, Haroutounian S, Kamerman P, et al. Neuropathic pain: an updated grading system for research and clinical practice. Pain. 2016;157(8):1599–1606.
  • Johnson MI. The Landscape of Chronic Pain: broader Perspectives. Medicina (Kaunas). 2019;55(5):1-19.
  • Sluka KA, Clauw DJ. Neurobiology of fibromyalgia and chronic widespread pain. Neuroscience. 2016;338:114–129.
  • Cohen SP, Mao J. Neuropathic pain: mechanisms and their clinical implications. Br Med J. 2014;348:f7656.
  • Orhurhu V, Orhurhu MS, Bhatia A, et al. Ketamine infusions for chronic pain: A systematic review and meta-analysis of randomized controlled trials. Anesth Analg. 2019;129(1):241–254.
  • Melzack RCKSensory, motivational, and central control determinants of pain: a new conceptual modelThe skin senses: proceedings of the first international symposium of the skin sensesKenshalo DeditorCharles C. ThomasFlorida1968Florida State University in Tallahassee,423–443.
  • Melzack R. The short-form mcgill pain questionnaire. Pain. 1987;30:191–197.
  • Cleeland CS, Ryan KM. Pain assessment: global use of the brief pain inventory. Ann Acad Med Singapore. 1994;23:129–138.
  • Ware JE Jr., Sherbourne CD. The MOS 36-item short-form health survey (SF-36). Conceptual framework and item selection. Med Care. 1992;30:473–483.
  • Uddin Z, MacDermid JC. Quantitative sensory testing in chronic musculoskeletal pain. Pain Med. 2016;17:1694–1703.
  • Hocking G, MJ C. Ketamine in chronic pain management: an evidence-based review. Anesth Analg. 2003;97:1730–1739.
  • Himmelseher S, Durieux ME. Ketamine for perioperative pain management. Anesthesiology. 2005;102:211–220.
  • Bell RF, Dahl JB, Moore RA, et al. Peri-operative ketamine for acute post-operative pain: a quantitative and qualitative systematic review (Cochrane review). Acta Anaesthesiol Scand. 2005;49:1405–1428.
  • Dahan A, Olofsen E, Sigtermans M, et al. Population pharmacokinetic-pharmacodynamic modeling of ketamine-induced pain relief of chronic pain. Eur J Pain. 2011;15:258–267.
  • Peltoniemi MA, Hagelberg NM, Olkkola KT, et al. Ketamine: A review of clinical pharmacokinetics and pharmacodynamics in anesthesia and pain therapy. Clin Pharmacokinet. 2016;55:1059–1077.
  • Leung LY, Baillie TA. Comparative pharmacology in the rat of ketamine and its two principal metabolites, norketamine and (Z)-6-hydroxynorketamine. J Med Chem. 1986;29:2396–2399.
  • Sigtermans MJ, van Hilten JJ, Bauer MC, et al. Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1. Pain. 2009;145:304–311.
  • Sigtermans M, Dahan A, Mooren R, et al. S(+)-ketamine effect on experimental pain and cardiac output: a population pharmacokinetic-pharmacodynamic modeling study in healthy volunteers. Anesthesiology. 2009;111:892–903.
  • Goldberg ME, Torjman MC, Schwartzman RJ, et al. Pharmacodynamic profiles of ketamine (R)- and (S)- with 5-day inpatient infusion for the treatment of complex regional pain syndrome. Pain Physician. 2010;13:379–387.
  • Herd DW, Anderson BJ, Holford NH. Modeling the norketamine metabolite in children and the implications for analgesia. Paediatr Anaesth. 2007;17:831–840.
  • J HM S, Voss L, Ketamine DB. More mechanisms of action than just NMDA blockade. Trends Anesthesia Crit Care. 2014;4:76–81.
  • Woolf CJ, Thompson SW. The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain. 1991;44:293–299.
  • 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;424:402–406.
  • 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;26:1235–1238.
  • Eide PK, Jorum E, Stubhaug A, et al. Relief of post-herpetic neuralgia with the N-methyl-D-aspartic acid receptor antagonist ketamine: a double-blind, cross-over comparison with morphine and placebo. Pain. 1994;58:347–354.
  • Eichenberger U, Neff F, Sveticic G, et al. Chronic phantom limb pain: the effects of calcitonin, ketamine, and their combination on pain and sensory thresholds. Anesth Analg. 2008;106:1265–1273.
  • 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;15:942–949.
  • Lodge D, Watkins JC, Bortolotto ZA, et al. The 1980s: D-AP5, LTP and a decade of NMDA receptor discoveries. Neurochem Res. 2019;44:516–530.
  • Okon T. Ketamine: an introduction for the pain and palliative medicine physician. Pain Physician. 2007;10:493–500.
  • Collingridge GL, Kehl SJ, McLennan H. Excitatory amino acids in synaptic transmission in the Schaffer collateral-commissural pathway of the rat hippocampus. J Physiol. 1983;334:33–46.
  • Cahusac PM, Evans RH, Hill RG, et al. The behavioural effects of an N-methylaspartate receptor antagonist following application to the lumbar spinal cord of conscious rats. Neuropharmacology. 1984;23:719–724.
  • Aanonsen LM, Wilcox GL. Nociceptive action of excitatory amino acids in the mouse: effects of spinally administered opioids, phencyclidine and sigma agonists. J Pharmacol Exp Ther. 1987;243:9–19.
  • Yang C, Yang J, Luo A, et al. Molecular and cellular mechanisms underlying the antidepressant effects of ketamine enantiomers and its metabolites. Transl Psychiatry. 2019;9(1):280.
  • Zanos P, Gould TD. Mechanisms of ketamine action as an antidepressant. Mol Psychiatry. 2018;23(4):801–811.
  • Garcia LS, Comim CM, Valvassori SS, et al. Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(1):140–144.
  • Lumsden EW, Troppoli TA, Myers SJ, et al. Antidepressant-relevant concentrations of the ketamine metabolite (2R,6R)-hydroxynorketamine do not block NMDA receptor function. Proc Nat Acad Sci. 2019;116(11):5160–5169.
  • Kavalali ET, Monteggia LM, Metabolite TK. 2R,6R-hydroxynorketamine blocks nmda receptors and impacts downstream signaling linked to antidepressant effects. Neuropsychopharmacology. 2018;43(1):221–222.
  • Rabben T, Oye I. Interindividual differences in the analgesic response to ketamine in chronic orofacial pain. Eur J Pain. 2001;5(3):233–240.
  • Li Y, Zhu ZR, Ou BC, et al. Dopamine D2/D3 but not dopamine D1 receptors are involved in the rapid antidepressant-like effects of ketamine in the forced swim test. Behav Brain Res. 2015;279:100–105.
  • Pacheco Dda F, Romero TR, Duarte ID. Central antinociception induced by ketamine is mediated by endogenous opioids and mu- and delta-opioid receptors. Brain Res. 2014;1562:69–75.
  • Fidecka S. Interactions of ketamine, naloxone and morphine in the rat. Pol J Pharmacol. 1987;39(1):33–40.
  • Campos AR, Santos FA, Rao VS. Ketamine-induced potentiation of morphine analgesia in rat tail-flick test: role of opioid-, alpha2-adrenoceptors and ATP-sensitive potassium channels. Biol Pharm Bull. 2006;29(1):86–89.
  • Mikkelsen S, Ilkjaer S, Brennum J, et al. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology. 1999;90(6):1539–1545.
  • Yanagihara Y, Ohtani M, Kariya S, et al. Plasma concentration profiles of ketamine and norketamine after administration of various ketamine preparations to healthy Japanese volunteers. Biopharm Drug Dispos. 2003;24(1):37–43.
  • Grant IS, Nimmo WS, Clements JA. Pharmacokinetics and analgesic effects of i.m. and oral ketamine. Br J Anaesth. 1981;53(8):805–810.
  • Chong C, Schug SA, Page-Sharp M, et al. Development of a sublingual/oral formulation of ketamine for use in neuropathic pain: preliminary findings from a three-way randomized, crossover study. Clin Drug Investig. 2009;29(5):317–324.
  • Fanta S, Kinnunen M, Backman JT, et al. Population pharmacokinetics of S-ketamine and norketamine in healthy volunteers after intravenous and oral dosing. Eur J Clin Pharmacol. 2015;71(4):441–447.
  • Carr DB, Goudas LC, Denman WT, et al. Safety and efficacy of intranasal ketamine for the treatment of breakthrough pain in patients with chronic pain: a randomized, double-blind, placebo-controlled, crossover study. Pain. 2004;108(1–2):17–27.
  • Giorgetti R, Marcotulli D, Tagliabracci A, et al. Effects of ketamine on psychomotor, sensory and cognitive functions relevant for driving ability. Forensic Sci Int. 2015;252:127–142.
  • Chaparro LE, Smith SA, Moore RA, et al. Pharmacotherapy for the prevention of chronic pain after surgery in adults. Cochrane Database Syst Rev. 2013;7:CD008307.
  • McNicol ED, Schumann R, Haroutounian S. A systematic review and meta-analysis of ketamine for the prevention of persistent post-surgical pain. Acta Anaesthesiol Scand. 2014;58:1199–1213.
  • Klatt E, Zumbrunn T, Bandschapp O, et al. Intra- and postoperative intravenous ketamine does not prevent chronic pain: A systematic review and meta-analysis. Scand J Pain. 2015;7:42–54.
  • Schwartzman RJ, Alexander GM, Grothusen JR, et al. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain. 2009;147:107–115.
  • Orhurhu VJ, Roberts JS, Cohen SP. Ketamine in acute and chronic pain management. StatPearls. Tampa/St. Petersburg, FL: Treasure Island StatPearls Publishing; 2019.
  • Bell RF, Dahl JB, Moore RA, et al. Peri-operative ketamine for acute post-operative pain: a quantitative and qualitative systematic review (Cochrane review). Acta Anaesthesiol Scand. 2005;49(10):1405–1428.
  • Anwar S, Cooper J, Rahman J, et al. Prolonged perioperative use of pregabalin and ketamine to prevent persistent pain after cardiac surgery. Anesthesiology. 2019;131:119–131.
  • Schwenk ES, Goldberg SF, Patel RD, et al. Adverse drug effects and preoperative medication factors related to perioperative low-dose ketamine infusions. Reg Anesth Pain Med. 2016;41:482–487.
  • Cohen SP, Wang S, Chen L, et al. An intravenous ketamine test as a predictive response tool in opioid-exposed patients with persistent pain. J Pain Symptom Manage. 2009;37:698–708.
  • Cohen SP, Verdolin MH, Chang AS, et al. The intravenous ketamine test predicts subsequent response to an oral dextromethorphan treatment regimen in fibromyalgia patients. J Pain. 2006;7:391–398.
  • Cohen SP, Chang AS, Larkin T, et al. The intravenous ketamine test: a predictive response tool for oral dextromethorphan treatment in neuropathic pain. Anesth Analg. 2004;99:1753–1759.
  • Elia N, Tramer MR. Ketamine and postoperative pain–a quantitative systematic review of randomised trials. Pain. 2005;113:61–70.
  • Noppers IM, Niesters M, Aarts LP, et al. Drug-induced liver injury following a repeated course of ketamine treatment for chronic pain in CRPS type 1 patients: a report of 3 cases. Pain. 2011;152:2173–2178.
  • Wong GL, Tam YH, Ng CF, et al. Liver injury is common among chronic abusers of ketamine. Clin Gastroenterol Hepatol. 2014;12:1759–62.e1.
  • Morgan CJ, Curran HV. Ketamine use: a review. Addiction. 2012;107:27–38.
  • Gregoire MC, MacLellan DL, Finley GA. A pediatric case of ketamine-associated cystitis. Urology. 2008;71:1232–1233.
  • Pagel PS, Kampine JP, Schmeling WT, et al. Ketamine depresses myocardial contractility as evaluated by the preload recruitable stroke work relationship in chronically instrumented dogs with autonomic nervous system blockade. Anesthesiology. 1992;76:564–572.
  • Ward J, Standage C. Angina pain precipitated by a continuous subcutaneous infusion of ketamine. J Pain Symptom Manage. 2003;25:6–7.
  • Tajerian M, Leu D, Yang P, et al. Differential efficacy of ketamine in the acute versus chronic stages of complex regional pain syndrome in mice. Anesthesiology. 2015;123(6):1435–1447.
  • Liman S, Cheung CW, Wong KL, et al. Preventive treatment with ketamine attenuates the ischaemia-reperfusion response in a chronic postischaemia pain model. Oxid Med Cell Longev. 2015;2015:380403.
  • Sasaki A, Serizawa K, Andoh T, et al. Pharmacological differences between static and dynamic allodynia in mice with herpetic or postherpetic pain. J Pharmacol Sci. 2008;108(3):266–273.
  • Zhou H, Zhang Q, Martinez E, et al. Ketamine reduces aversion in rodent pain models by suppressing hyperactivity of the anterior cingulate cortex. Nat Commun. 2018;9(1):3751.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.