6,164
Views
37
CrossRef citations to date
0
Altmetric
Review

Pharmacokinetic and pharmacodynamic considerations for NMDA-receptor antagonist ketamine in the treatment of chronic neuropathic pain: an update of the most recent literature

, , , ORCID Icon, ORCID Icon &
Pages 1033-1041 | Received 22 Aug 2019, Accepted 04 Nov 2019, Published online: 13 Nov 2019

References

  • International Association for the Study of Pain (IASP). IASP terminology: neuropathic pain. Available from: https://www.iasp-pain.org/Education/Content.aspx?ItemNumber=1698#Neuropathicpain
  • Scholz J, Finnerup NB, Attal N, et al. The IASP classification of chronic pain for ICD-11. Pain. 2019;160:53–59.
  • Jensen TS, Finnerup NB. Allodynia and hyperalgesia in neuropathic pain: clinical manifestations and mechanisms. Lancet Neurol. 2014;13:924–935.
  • Arendt-Nielsen L, Morlion B, Perrot S, et al. Assessment and manifestation of central sensitization across different chronic pain conditions. Eur J Pain. 2018;22:216–241.
  • Dahan A, van Velzen M, Niesters M. Comorbidities and the complexities of chronic pain. Anesthesiology. 2014;121:1–3.
  • Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14:162–173.
  • Niesters M, Dahan A. Pharmacokinetic and pharmacodynamic considerations for NMDA receptor antagonists in the treatment of chronic neuropathic pain. Exp Opin Drug Metab. 2012;8:1409–1417.
  • Niesters M, Martini C, Dahan A. Ketamine for chronic pain: risks and benefits. Br J Clin Pharmacol. 2014;77:357–367.
  • Jonkman K, Dahan A, van de Donk T, et al. Ketamine for pain. F1000Res. 2017;6:1711.
  • Collins S, Sigtermans M, Dahan A, et al. NMDA receptor antagonists for the treatment of neuropathic pain. Pain Med. 2010;11:1726–1742.
  • Aiyer R, Mehta N, Gungor S, et al. A systematic review of NMDA receptor antagonists for treatment of neuropathic pain in clinical practice. Clin J Pain. 2018;34:450–467.
  • Arendt-Nielsen L, Nielsen J, Petersen-Felix S, et al. Effect of racemic ketamine and the (S+)-isomer of ketamine on temporal and spatial summation of pain. Br J Anaesth. 1996;77:625–631.
  • Bosma RL, Cheng JC, Rogachov A, et al. Brain dynamics and temporal summation of pain predicts neuropathic pain relief from ketamine infusion. Anesthesiology. 2018;129:1015–1024.
  • Zanos P, Moaddel R, Morris PJ. et al. NMDAR inhibition-independent antidepressant action of ketamine metabolism. Nature. 2016;533:481–486.
  • Liriano F, Hatten C, Schwartz TL. Ketamine as treatment for post-traumatic stress disorder: a review. Drugs Context. 2019;8:212305.
  • Rehder KJ. Adjunct therapies for refractory status asthmaticus in children. Respir Care. 2017;62:849–865.
  • Dale O, Somogyi AA, Li Y, et al. Does intraoperative ketamine attenuate the inflammatory reactivity following surgery? A systematic review and meta-analysis. Anesth Analg. 2012;115:934–943.
  • Swartjes M, Niesters M, Heij L, et al. Ketamine does not produce relief of neuropathic pain in mice lacking the β-common receptor (CD131). PLoS One. 2013;8:e71326.
  • Jonkman K, van der Schrier R, van Velzen M, et al. Psychedelic symptoms induced by racemic ketamine and esketamine are differently reduced by nitric oxide donor sodium nitroprusside. A proof-of-concept study in healthy male volunteers. Br J Anaesth. 2018;120:1009–1018.
  • Jonkman K, Duma A, Olofsen E, et al. Pharmacokinetics and bioavailability of inhaled esketamine in healthy volunteers. Anesthesiology. 2017;127:675–683.
  • Nielsen BN, Friis SM, Romsing J, et al. Intranasal sufentanil/ketamine analgesia in children. Paediatr Anaesth. 2014;24:170–180.
  • https://www.fda.gov/news-events/press-announcements/fda-approves-new-nasal-spray-medication-treatment-resistant-depression-available-only-certified
  • Kaufman MB. Pharmaceutical approval update. Pharm Ther. 2019;44:252.
  • Kamp J, Olofsen E, Henthorn TK, et al. Pharmacokinetic of the N-methyl-D-aspartate receptor antagonist ketamine: a systematic review of the literature. Submitted
  • Zanos P, Moaddel R, Morris PJ, et al. Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharm Rev. 2018;70:621–660.
  • Hijazi Y, Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30:853–858.
  • Noppers I, Olofsen E, Niesters M, et al. Effect of rifampicin on S-ketamine and S-norketamine plasma concentrations in healthy volunteers after intravenous S-ketamine administration. Anesthesiology. 2011;114:1435–1445.
  • Yanagihara Y, Kariya S, Ohtani M, et al. Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2001;29:887–890.
  • Ashraf MW, Peltoniemi MA, Olkkola KT, et al. Semi-mechanistic population pharmacokinetic model to predict the drug-drug interaction between S-ketamine and ticlopidine in healthy human volunteers. CPT Pharmacometrics Syst Pharmacol. 2018;10:687–697.
  • Li Y, Jackson KA, Slon B, et al. CYP2B6*6 allele and age substantially reduce steady-state ketamine clearance in chronic pain patients: impact on adverse effects. Br J Clin Pharmacol. 2015;80:276–284.
  • Zhao X, Venkata SL, Moaddel R, et al. Simultaneous population pharmacokinetic modelling of ketamine and three major metabolites in patients with treatment-resistant bipolar depression. Br J Clin Pharmacol. 2012;74:304–314.
  • Henthorn TK, Avram MJ, Dahan A, et al. Combined recirculatory-compartmental population pharmacokinetic modeling of arterial and venous plasma S(+)- and R(-)-ketamine concentrations. Anesthesiology. 2018;129:260–270.
  • Moaddel R, Sanghvi M, Dossou KSS, et al. The distribution and clearance of (2S,6S)-hydroxynorketamine, an active ketamine metabolite, in Wistar rats. Pharm Res Per. 2015;3:e005157.
  • 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;129:241–254.
  • Zhao J, Wang Y, Wang D. The effect of ketamine infusion in the treatment of complex regional pain syndrome: a systematic review and meta-analysis. Curr Pain Headache Rep. 2018;22:12.
  • Michelet D, Brasher C, Horlin AL. et al. Ketamine for chronic non-cancer pain: a meta-analysis and trial sequential analysis of randomized trials. Eur J Pain. 2018;22:632–646.
  • Connolly SB, Prager JP, Norman HR. A systematic review of ketamine in complex regional pain syndrome. Pain Med. 2015;16:943–969.
  • Bell RF, Eccleston C, Kalso EA. Ketamine as adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2017;6:CD003351.
  • Jonkman K, van de Donk T, Dahan A. Ketamine for cancer pain: what is the evidence? Curr Opin Support Palliat Care. 2017;11:88–92.
  • Fallon MT, Wilcock A, Kelly CA, et al. Oral ketamine vs placebo in patients with cancer-related neuropathic pain: a randomized clinical trial. JAMA Oncol. 2018;4:870–872.
  • Nielsen RV, Fomsgaard JS, Siegel H, et al. Intraoperative ketamine reduces immediate postoperative opioid consumption after spinal fusion surgery in chronic pain patients with opioid dependency: a randomized, blinded trial. Pain. 2017;158:463–470.
  • Loftus RW, Yeager MP, Clark JA, et al. Intraoperative ketamine reduces perioperative opiate consumption in opiate-dependent patients with chronic back pain undergoing surgery. Anesthesiology. 2010;113:639–646.
  • Nielsen VR, Fomsgaard JS, Nikolajsen L, et al. Intraoperative S-ketamine for the reduction of opioid consumption and pain one year after spine surgery: a randomized clinical trial of opioid-dependent patients. Eur J Pain. 2019;23:455–460.
  • Ocker AC, Shah NB, Schwenk ES. et al. Ketamine and cognitive behavioral therapy for rapid opioid tapering with sustained opioid abstinence: a case report and 1-year follow-up. Pain Pract. 2019; Epub ahead of print
  • Kroin JS, Das V, Moric M, et al. Efficacy of the ketamine metabolite (2R,6R)-hydroxynorketamine in mice models of pain. Reg Anesth Pain Med. 2019;44:111–117.
  • Swartjes M, Morariu A, Niesters M, et al. Non-selective and NR2B-selective NMDA receptor antagonists produce antinociception and long-term relief of allodynia in acute and neuropathic pain. Anesthesiology. 2011;115:165–174.
  • Niesters M, Khalili-Mahani N, Martini C, et al. Effect of sub-anesthetic ketamine on intrinsic functional brain connectivity: a placebo-controlled functional magnetic resonance imaging study in healthy volunteers. Anesthesiology. 2012;117:868–877.
  • Maia-de-Oliveira JP, Lobão-Soares B, Ramalho T, et al. Nitroprusside single-dose prevents the psychosis-like behavior induced by ketamine in rats for up to one week. Schizophr Res. 2015;162:211–215.
  • Bujas-Bobanovic M, Bird DC, Robertson HA, et al. Blockade of phencyclidine-induced effects by nitric oxide donor. Br J Pharmacol. 2000;130:1005–1012.
  • Treviopoulou A, Touzlatzi N, Pitsikas N. The nitric oxide donor sodium nitroprusside attenuates recognition memory deficits and social withdrawal produced by the NMDA receptor antagonist ketamine and induces anxiolytic-like behavior in rats. Psychopharmacol. 2016;233:1045–1054.
  • Bird DC, Bujas-Bobanovic M, Robertson HA, et al. Lack of phencyclidine-induced effect in mice with reduced neuronal nitric oxide synthase. Psychopharmacol. 2011;155:299–309.
  • Hallak JEC, Maia-de-Oliveira JP, Abrao J, et al. Rapid improvement of acute schizophrenia symptoms after intravenous sodium nitroprusside: a randomized, double-blind, placebo-controlled trial. JAMA Psychiatry. 2013;70:668–676.
  • Luckenbaugh DA, Niciu MJ, Ionescu DF, et al. Do the dissociative side effects of ketamine mediate its antidepressant effects? J Affect Dis. 2004;159:56–61.
  • Sigtermans MJ, van Hilten JJ, Beuer MCR, et al. Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1. Pain. 2009;145:304–311.
  • Borsook D. Ketamine and chronic pain – going the distance. Pain. 2009;145:271–272.
  • MacKintosh D. Ketamine and cancer pain – an inconvenient truth? Ann Palliat Med. 2012;1:224–226.
  • Oudejans LCJ, van Velzen M, Olofsen E, et al. Translation of random painful stimuli into numerical responses in fibromyalgia and perioperative patients. Pain. 2016;157:128–136.
  • Torensma, B, Oudejans L, van Velzen M, et al. Pain sensitivity and pain scoring in patients with morbid obesity. SOARD. 2017;13(5):788–795.
  • Tuttle AH, Tohyama S, Ramsay T, et al. Increasing placebo responses over time in U.S. clinical trials of neuropathic pain. Pain. 2015;156:2616–2626.
  • Ledford H. A question of control. Nature. 2018;563:312–315.
  • Woodcock J, LaVange LM. Master protocols to study multiple therapies, multiple diseases, or both. N Engl J Med. 2017;377:62–70.
  • Okie A. A flood of opioids, a rising tide of deaths. N Engl J Med. 2010;363:1981–1985.
  • Bedene A, Lijfering WM, Niesters M, et al. Opioid prescription patterns and risk factors associated with opioid use in the Netherlands. JAMA Network Open. 2019;2(8):e1910223.
  • Jonkman K, van Rijnsoever E, Olofsen E, et al. Esketamine counters opioid-induced respiratory depression. Br J Anaesth. 2018;120:1117–1127.
  • Dahan A, van der Schrier R, Smith T, et al. Averting opioid-induced respiratory depression without affecting analgesia. Anesthesiology. 2018;128:1027–1037.