1,382
Views
76
CrossRef citations to date
0
Altmetric
Review

An update on ketamine and its two enantiomers as rapid-acting antidepressants

&
Pages 83-92 | Received 10 Sep 2018, Accepted 28 Nov 2018, Published online: 04 Dec 2018

References

  • Ebert B, Mikkelsen S, Thorkildsen C, et al. Norketamine, the main metabolite of ketamine, is a non-competitive NMDA receptor antagonist in the rat cortex and spinal cord. Eur J Pharmacol. 1997;333(1):99–104.
  • Domino EF. Taming the ketamine tiger. Anesthesiology. 2010;113(3):678–684.
  • Tyler MW, Yourish HB, Ionescu DF, et al. Classics in chemical neuroscience: ketamine. ACS Chem Neurosci. 2017;8(6):1122–1134.
  • Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47(4):351–354.
  • Duman RS. Ketamine and rapid-acting antidepressants: a new era in the battle against depression and suicide. F1000Res. 2018;7(F1000Faculty Rev):659.
  • Zarate CA, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856–864.
  • Price RB, Nock MK, Charney DS, et al. Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry. 2009;66(5):522–526.
  • Larkin GL, Beautrais AL. A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol. 2011;14(8):1127–1131.
  • Su TP, Chen MH, Li CT, et al. Dose-related effects of adjunctive ketamine in Taiwanese patients with treatment-resistant depression. Neuropsychopharmacology. 2017;42(13):2482–2492.
  • Murrough JW, Iosifescu DV, Chang LC, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry. 2013;170(10):1134–1142.
  • Murrough JW, Soleimani L, DeWilde KE, et al. Ketamine for rapid reduction of suicidal ideation: a randomized controlled trial. Psychol Med. 2015;45(16):3571–3580.
  • Grunebaum MF, Galfalvy HC, Choo TH, et al. Ketamine for rapid reduction of suicidal thoughts in major depression: a midazolam-controlled randomized clinical trial. Am J Psychiatry. 2018;175(4):327–335.
  • Frye MA, Blier P, Tye SJ. Concomitant benzodiazepine use attenuates ketamine response: implications for large scale study design and clinical development. J Clin Psychopharmacol. 2015;35(3):334–336.
  • Albot CS, Shiroma PR, Cullen KR, et al. The antidepressant effect of repeat dose intravenous ketamine is delayed by concurrent benzodiazepine use. J Clin Psychiatry. 2017;78(3):e308–e309.
  • Aan Het Rot M, Ca Z, Ds C, et al. Ketamine for depression: where do we go from here? Biol Psychiatry. 2012;72(1):537–547.
  • Aan Het Rot M, Collins KA, Murrough JW, et al. Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry. 2010;67(2):139–145.
  • Murrough JW, Perez AM, Pillemer S, et al. Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry. 2013;74(4):250–256.
  • Singh JB, Fedgchin M, Daly EJ, et al. A double-blind, randomized, placebo-controlled, dose-frequency study of intravenous ketamine in patients with treatment-resistant depression. Am J Psychiatry. 2016;173(8):816–826.
  • Li L, Vlisides PE. Ketamine: 50 years of modulating the mind. Front Hum Neurosci. 2016;10:612.
  • 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(9):1059–1077.
  • Loo CK, Gálvez V, O’keefe E, et al. Placebo‐controlled pilot trial testing dose titration and intravenous, intramuscular and subcutaneous routes for ketamine in depression. Acta Psychiatr Scand. 2016;134(1):48–56.
  • Lapidus KAB, Levitch CF, Perez AM, et al. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol Psychiatry. 2014;76(12):970–976.
  • Irwin SA, Iglewicz A, Nelesen RA, et al. Daily oral ketamine for the treatment of depression and anxiety in patients receiving hospice care: a 28-day open-label proof-of-concept trial. J Palliat Med. 2013;16(8):958–965.
  • Lara DR, Bisol LW, Munari LR. Antidepressant, mood stabilizing and procognitive effects of very low dose sublingual ketamine in refractory unipolar and bipolar depression. Int J Neuropsychopharmacol. 2013;16(9):2111–2117.
  • Diazgranados N, Ibrahim L, Brutsche NE, et al. A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry. 2010;67(8):793–802.
  • Zarate Jr CA, Brutsche NE, Ibrahim L, et al. Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry. 2012;71(11):939–946.
  • Nugent AC, Diazgranados N, Carlson PJ, et al. Neural correlates of rapid antidepressant response to ketamine in bipolar disorder. Bipolar Disord. 2014;16(2):119–128.
  • Viktorin A, Lichtenstein P, Thase ME, et al. The risk of switch to mania in patients with bipolar disorder during treatment with an antidepressant alone and in combination with a mood stabilizer. Am J Psychiatry. 2014;171(10):1067–1073.
  • Romeo B, Choucha W, Fossati P, et al. Meta-analysis of short-and mid-term efficacy of ketamine in unipolar and bipolar depression. Psychiatry Res. 2015;230(2):682–688.
  • Niciu MJ, Luckenbaugh DA, Ionescu DF, et al. Subanesthetic dose ketamine does not induce an affective switch in three independent samples of treatment-resistant major depression. Biol Psychiatry. 2013;74(10):e23–e24.
  • Wilkinson ST, Toprak M, Turner MS, et al. A survey of the clinical, off-label use of ketamine as a treatment for psychiatric disorders. Am J Psychiatry. 2017;174(7):695–696.
  • Galea S, Nandi A, Vlahov D. The epidemiology of post-traumatic stress disorder after disasters. Epidemiol Rev. 2005;27(1):78–91.
  • McGhee LL, Maani CV, Garza TH, et al. The correlation between ketamine and posttraumatic stress disorder in burned service members. J Trauma Acute Care Surg. 2008;64(2):S195–S199.
  • Brachman RA, McGowan JC, Perusini JN, et al. Ketamine as a prophylactic against stress-induced depressive-like behavior. Biol Psychiatry. 2016;79(9):776–786.
  • Mastrodonato A, Martinez R, Pavlova IP, et al. Ventral CA3 activation mediates prophylactic ketamine efficacy against stress-induced depressive-like behavior. Biol Psychiatry. 2018;84(11):846-856.
  • 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(6):681–688.
  • Hartberg J, Garrett-Walcott S, De Gioannis A. Impact of oral ketamine augmentation on hospital admissions in treatment-resistant depression and PTSD: a retrospective study. Psychopharmacology (Berl). 2018;235(2):393–398.
  • Rodriguez CI, Kegeles LS, Flood P, et al. Rapid resolution of obsessions after an infusion of intravenous ketamine in a patient with treatment-resistant obsessive-compulsive disorder: a case report. J Clin Psychiatry. 2011;72(4):567.
  • Rodriguez CI, Kegeles LS, Levinson A, et al. Randomized controlled crossover trial of ketamine in obsessive-compulsive disorder: proof-of-concept. Neuropsychopharmacology. 2013;38(12):2475.
  • Rodriguez CI, Wheaton M, Zwerling J, et al. Can exposure-based CBT extend IV ketamine’s effects in obsessive-compulsive disorder? An open-label trial. J Clin Psychiatry. 2016;7(3):408.
  • Bloch MH, Wasylink S, Landeros-Weisenberger A, et al. Effects of ketamine in treatment-refractory obsessive-compulsive disorder. Biol Psychiatry. 2012;72(11):964–970.
  • Glue P, Neehoff SM, Medlicott NJ, et al. Safety and efficacy of maintenance ketamine treatment in patients with treatment-refractory generalised anxiety and social anxiety disorders. J Psychopharmacol. 2018;32(6):663–667.
  • Taylor JH, Landeros-Weisenberger A, Coughlin C, et al. Ketamine for social anxiety disorder: a randomized, placebo-controlled crossover trial. Neuropsychopharmacology. 2018;43(2):325–333.
  • Shadli SM, Kawe T, Martin D, et al. Ketamine effects on EEG during therapy of treatment-resistant generalized anxiety and social anxiety. Int J Neuropsychopharmacol. 2018;8(1):717–724.
  • Laje G, Lally N, Mathews D, et al. Brain-derived neurotrophic factor Val66Met polymorphism and antidepressant efficacy of ketamine in depressed patients. Biol Psychiatry. 2012;72(11):e27–e28.
  • Hashimoto K. A BDNF Val66Met polymorphism and ketamine-induced rapid antidepressant action. Clin Psychopharmacol Neurosci. 2012;10(1):59–60.
  • Yang JJ, Wang N, Yang C, et al. Serum interleukin-6 is a predictive biomarker for ketamine’s antidepressant effect in treatment-resistant patients with major depression. Biol Psychiatry. 2015;77(3):e19–e20.
  • Kiraly DD, Horn SR, Van Dam NT, et al. Altered peripheral immune profiles in treatment-resistant depression: response to ketamine and prediction of treatment outcome. Transl Psychiatry. 2017;7(3):e1065.
  • Park M, Newman LE, Gold PW, et al. Change in cytokine levels is not associated with rapid antidepressant response to ketamine in treatment-resistant depression. J Psychiatr Res. 2017;84:113–118.
  • Chen MH, Li CT, Lin WC, et al. Rapid inflammation modulation and antidepressant efficacy of a low-dose ketamine infusion in treatment-resistant depression: a randomized, double-blind control study. Psychiatry Res. 2018;269:207–211.
  • Kadriu B, Gold PW, Luckenbaugh DA, et al. Acute ketamine administration corrects abnormal inflammatory bone markers in major depressive disorder. Mol Psychiatry. 2018;23:1626–1631.
  • Zhang K, Ma M, Dong C, et al. Role of inflammatory bone markers in the antidepressant actions of (R)-ketamine in a chronic social defeat stress model. Int J Neuropsychopharmacol. 2018;21(11):1025-1030.
  • Moaddel R, Shardell M, Khadeer M, et al. Plasma metabolomic profiling of a ketamine and placebo crossover trial of major depressive disorder and healthy control subjects. Psychopharmacology (Berl). 2018;235(10):3017–3030.
  • Li N, Lee B, R J L, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959–964.
  • Hashimoto K. Role of the mTOR signaling pathway in the rapid antidepressant action of ketamine. Expert Rev Neurother. 2011;11(1):33–36.
  • Denk MC, Rewerts C, Holsboer F, et al. Monitoring ketamine treatment response in a depressed patient via peripheral mammalian target of rapamycin activation. Am J Psychiatry. 2011;168(7):751–752.
  • Yang C, Zhou Z, Gao Z, et al. Acute increases in plasma mammalian target of rapamycin, glycogen synthase kinase-3β, and eukaryotic elongation factor 2 phosphorylation after ketamine treatment in three depressed patients. Biol Psychiatry. 2013;73(12):e35–e36.
  • Yang C, Ren Q, Qu Y, et al. Mechanistic target of rapamycin-independent antidepressant effects of (R)-ketamine in a social defeat stress model. Biol Psychiatry. 2018;83(1):18–28.
  • Singh JB, Fedgchin M, Daly E, et al. Intravenous esketamine in adult treatment-resistant depression: a double-blind, double-randomization, placebo-controlled study. Biol Psychiatry. 2016;80(6):424–431.
  • Correia-Melo FS, Argolo FC, Araújo-de-Freitas L, et al. Rapid infusion of esketamine for unipolar and bipolar depression: a retrospective chart review. Neuropsychiatr Dis Treat. 2017;13:1627–1632.
  • Bartova L, Papageorgiou K, Milenkovic I, et al. Rapid antidepressant effect of S-ketamine in schizophrenia. Eur Neuropsychopharmacol. 2018 Aug;28(8):980–982.
  • Morrison RL, Fedgchin M, Singh J, et al. Effect of intranasal esketamine on cognitive functioning in healthy participants: a randomized, double-blind, placebo-controlled study. Psychopharmacology (Berl). 2018;235(4):1107–1119.
  • Van de Loo AJAE, Bervoets AC, Mooren L, et al. The effects of intranasal esketamine (84 mg) and oral mirtazapine (30 mg) on on-road driving performance: a double-blind, placebo-controlled study. Psychopharmacology (Berl). 2017;234(21):3175–3183.
  • Daly EJ, Singh JB, Fedgchin M, et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: a randomized clinical trial. JAMA Psychiatry. 2018;75(2):139–148.
  • Canuso CM, Singh JB, Fedgchin M, et al. Efficacy and safety of intranasal esketamine for the rapid reduction of symptoms of depression and suicidality in patients at imminent risk for suicide: results of a double-blind, randomized, placebo-controlled study. Am J Psychiatry. 2018;175(7):620–630.
  • 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(10):950–966.
  • Kishimoto T, Chawia JM, Hagi K, et al. Single-dose infusion ketamine and non-ketamine N-methyl-D-aspartate receptor antagonists for unipolar and bipolar depression: a meta-analysis of efficacy, safety and time trajectories. Psychol Med. 2016;46(7):1459–1472.
  • Papadimitropoulou K, Vossen C, Karabis A, et al. Comparative efficacy and tolerability of pharmacological and somatic interventions in adult patients with treatment-resistant depression: a systematic review and network meta-analysis. Curr Med Res Opin. 2017;33(4):701–711.
  • Kokkinou M, Ashok AH, Howes OD. The effects of ketamine on dopaminergic function: meta-analysis and review of the implications for neuropsychiatric disorders. Mol Psychiatry. 2018;23(1):59–69.
  • Zhang J, Li S, Hashimoto K. R (−)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacol Biochem Behav. 2014;116:137–141.
  • Yang C, Shirayama Y, Zhang JC, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5(9):e632.
  • Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533(7604):481–486.
  • Fukumoto K, Toki H, Iijima M, et al. Antidepressant potential of (R)-ketamine in rodent models: comparison with (S)-ketamine. J Pharmacol Exp Ther. 2017;361(1):9–16.
  • Yang C, Qu Y, Abe M, et al. (R)-ketamine shows greater potency and longer lasting antidepressant effects than its metabolite (2R,6R)-hydroxynorketamine. Biol Psychiatry. 2017;82(5):e43–e44.
  • Yang C, Qu Y, Fujita Y, et al. Possible role of gut-microbiota in the antidepressant effects of (R)-ketamine in a social defeat stress model. Transl Psychiatry. 2017;7(12):1294.
  • Yang C, Han M, Zhang JC, et al. Loss of parvalbumin-immunoreactivity in mouse brain regions after repeated intermittent administration of esketamine, but not R-ketamine. Psychiatric Res. 2016;239:281–283.
  • Hashimoto K, Kakiuchi T, Ohba H, et al. Reduction of dopamine D2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys. Eur Arch Psychiatry Clin Neurosci. 2017;267(2):173–176.
  • Tian Z, Dong C, Fujita A, et al. Expression of heat shock protein HSP-70 in the retrosplenial cortex of rat brain after administration of (R,S)-ketamine and (S)-ketamine, but not (R)-ketamine. Pharmacol Biochem Behav. 2018;172:17–21.
  • Hashimoto K. The R-stereoisomer of ketamine as an alternative for ketamine for treatment-resistant major depression. Clin Psychopharmacol Neurosci. 2014;12(1):72–73.
  • Hashimoto K. R-ketamine: a rapid-onset and sustained antidepressant without risk of brain toxicity. Psychol Med. 2016;46(11):2449–2451.
  • Hashimoto K. Ketamine’s antidepressant action: beyond NMDA receptor inhibition. Expert Opin Ther Targets. 2016;20(11):1389–1392.
  • Hashimoto K. Detrimental side effects of repeated ketamine infusions in the brain. Am J Psychiatry. 2016;173(10):1044–1045.
  • Hashimoto K. Rapid-antidepressant activity of ketamine: beyond NMDA receptor. In: Hashimoto K, editor. The NMDA Receptors. New York: Humana Press; 2017. p. 69–82.
  • White PF, Schüttler J, Shafer A, et al. Comparative pharmacology of the ketamine isomers. Studies in volunteers. Br J Anaesth. 1985;57(2):197–203.
  • Vollenweider FX, Leenders KL, Oye I, et al. Differential psychopathology and patterns of cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using positron emission tomography (PET). Eur Neuropsychopharmacol. 1997;7(1):25–38.
  • Yang C, Kobayashi S, Nakao K, et al. AMPA receptor activation-independent antidepressant actions of ketamine metabolite (S)-norketamine. Biol Psychiatry. 2018;84(8):591–600.
  • Hashimoto K, Yang C. Is (S)-norketamine an alternative antidepressant for esketamine? Eur Arch Psychiatry Clin Neurosci. 2018 Jul 14. Doi:10.1007/s00406-018-0922-2.
  • Singh I, Morgan C, Curran V, et al. Ketamine treatment for depression: opportunities for clinical innovation and ethical foresight. Lancet Psychiatry. 2017;4(5):419–426.
  • Short B, Fong J, Galvez V, et al. Side-effects associated with ketamine use in depression: a systematic review. Lancet Psychiatry. 2018;5(1):65–78.
  • Shirayama Y, Hashimoto K. Effects of a single bilateral infusion of R-ketamine in the rat brain regions of a learned helplessness model of depression. Eur Arch Psychiatry Clin Neurosci. 2017;267(2):177–182.
  • Shirayama Y, Hashimoto K. Lack of antidepressant effects of (2R,6R)-hydroxynorketamine in a rat learned helplessness model: comparison with (R)-ketamine. Int J Neuropsychopharmacol. 2018;21(1):84–88.
  • Zhang K, Fujita Y, Hashimoto K. Lack of metabolism in (R)-ketamine’s antidepressant actions in a chronic social defeat stress model. Sci Rep. 2018;8(1):4007.
  • Hashimoto K, Shirayama Y. What are the causes for discrepancies of antidepressant actions of (2R,6R)-hydroxynorketamine? Biol Psychiatry. 2018;84(1):e7–e8.
  • Chaki S. Is metabolism of (R)-ketamine essential for the antidepressant effects?. Int J Neuropsychopharmacol. 2018;21(2):154–156.
  • Yamaguchi JI, Toki H, Qu Y, et al. (2R,6R)-Hydroxynorketamine is not essential for the antidepressant actions of (R)-ketamine in mice. Neuropsychopharmacology. 2018;43(9):1900–1907.
  • Chang L, Toki H, Qu Y, et al. No sex-specific differences in the acute antidepressant actions of (R)-ketamine in an inflammation model. Int J Neuropsychopharmacol. 2018;21(10):932–937.
  • Zhang K, Toki H, Fujita Y, et al. Lack of deuterium isotope effects in the antidepressant effects of (R)-ketamine in a chronic social defeat stress model. Psychopharmacology (Berl). 2018;235(11):3177–3185.

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.