3,643
Views
64
CrossRef citations to date
0
Altmetric
Drug Profile

Esketamine for treatment resistant depression

, , , , , , , , & show all
Pages 899-911 | Received 11 Apr 2019, Accepted 03 Jul 2019, Published online: 16 Jul 2019

References

  • Patten SB, Williams JVA, Lavorato DH, et al. Descriptive epidemiology of major depressive disorder in Canada in 2012. Can J Psychiatry. 2015;60:23–30.
  • Ustün TB, Ayuso-Mateos JL, Chatterji S, et al. Global burden of depressive disorders in the year 2000. Br J Psychiatry. 2004;184:386–392.
  • Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163:1905–1917.
  • McIntyre RS, Filteau M-J, Martin L, et al. Treatment-resistant depression: definitions, review of the evidence, and algorithmic approach. J Affect Disord. 2014;156:1–7.
  • Cleare A, Pariante CM, Young AH, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: A revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol (Oxford). 2015;29:459–525.
  • Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 3. pharmacological treatments. Can J Psychiatry. 2016;61:540–560.
  • Folkerts HW, Michael N, Tölle R, et al. Electroconvulsive therapy vs. paroxetine in treatment-resistant depression — a randomized study. Acta Psychiatrica Scand. 1997;96:334–342.
  • UK ECT Review Group. Efficacy and safety of electroconvulsive therapy in depressive disorders: a systematic review and meta-analysis. Lancet. 2003;361:799–808.
  • Berlim MT, McGirr A, Van den Eynde F, et al. Effectiveness and acceptability of deep brain stimulation (DBS) of the subgenual cingulate cortex for treatment-resistant depression: a systematic review and exploratory meta-analysis. J Affect Disord. 2014;159:31–38.
  • Martin JLR, Martín-Sánchez E. Systematic review and meta-analysis of vagus nerve stimulation in the treatment of depression: variable results based on study designs. Eur Psychiatry. 2012;27:147–155.
  • Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38:651–661.
  • Berlim MT, Van Den Eynde F, Jeff Daskalakis Z. Clinically meaningful efficacy and acceptability of low-frequency repetitive transcranial magnetic stimulation (rTMS) for treating primary major depression: a meta-analysis of randomized, double-blind and sham-controlled trials. Neuropsychopharmacology. 2013;38:543–551.
  • Berlim MT, van Den Eynde F, Tovar-Perdomo S, et al. Response, remission and drop-out rates following high-frequency repetitive transcranial magnetic stimulation (rTMS) for treating major depression: a systematic review and meta-analysis of randomized, double-blind and sham-controlled trials. Psychol Med. 2014;44:225–239.
  • Cao X, Deng C, Su X, et al. Response and remission rates following high-frequency vs. low-frequency repetitive transcranial magnetic stimulation (rTMS) over right DLPFC for treating major depressive disorder (MDD): a meta-analysis of randomized, double-blind trials. Front Psychiatry. 2018;9:413.
  • Senova S, Cotovio G, Pascual-Leone A, et al. Durability of antidepressant response to repetitive transcranial magnetic stimulation: systematic review and meta-analysis. Brain Stimul. 2019;12:119–128.
  • Anis NA, Berry SC, Burton NR, et al. The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol. 1983;79:565–575.
  • Lodge D, Anis NA, Burton NR. Effects of optical isomers of ketamine on excitation of cat and rat spinal neurones by amino acids and acetylcholine. Neurosci Lett. 1982;29:281–286.
  • Miller OH, Moran JT, Hall BJ. Two cellular hypotheses explaining the initiation of ketamine’s antidepressant actions: direct inhibition and disinhibition. Neuropharmacology. 2016;100:17–26.
  • Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–354.
  • 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;74:250–256.
  • Coyle CM, Laws KR. The use of ketamine as an antidepressant: a systematic review and meta-analysis. Hum Psychopharmacol. 2015;30:152–163.
  • Thomas RK, Baker G, Lind J, et al. Rapid effectiveness of intravenous ketamine for ultraresistant depression in a clinical setting and evidence for baseline anhedonia and bipolarity as clinical predictors of effectiveness. J Psychopharmacol (Oxford). 2018;32:1110–1117.
  • Archer S, Chrenek C, Swainson J. Maintenance ketamine therapy for treatment-resistant depression. J Clin Psychopharmacol. 2018;38:380–384.
  • Lapidus KAB, Levitch CF, Perez AM, et al. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol Psychiatry. 2014;76:970–976.
  • Gálvez V, Li A, Huggins C, et al. Repeated intranasal ketamine for treatment-resistant depression - the way to go? Results from a pilot randomised controlled trial. J Psychopharmacol (Oxford). 2018;32:397–407.
  • Lee V, Archer S, Chrenek C, et al. A response to: repeated intranasal ketamine for treatment resistant depression: the way to go? Results from a pilot randomised controlled trial. J Psychopharmacol (Oxford). 2019;33:258–259.
  • Abdallah CG, Adams TG, Kelmendi B, et al. Ketamine’s mechanism of action: a path to rapid-acting antidepressants. Depress Anxiety. 2016;33:689–697.
  • Baker GB, Prior TI. Stereochemistry and drug efficacy and development: relevance of chirality to antidepressant and antipsychotic drugs. Ann Med. 2002;34:537–543.
  • Andrade C. Ketamine for depression, 3: does chirality matter? J Clin Psychiatry. 2017;78:e674–e677.
  • Muller J, Pentyala S, Dilger J, et al. Ketamine enantiomers in the rapid and sustained antidepressant effects. Ther Adv Psychopharmacol. 2016;6:185–192.
  • 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.
  • Pfenninger EG, Durieux ME, Himmelseher S. Cognitive impairment after small-dose ketamine isomers in comparison to equianalgesic racemic ketamine in human volunteers. Anesthesiology. 2002;96:357–366.
  • Singh JB. Antidepressant efficacy and dosing comparisons of ketamine enantiomers: response to Hashimoto. Am J Psychiatry. 2016;173:1045–1046.
  • White PF, Schüttler J, Shafer A, et al. Comparative pharmacology of the ketamine isomers. Studies in volunteers. Br J Anaesth. 1985;57:197–203.
  • 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;361:9–16.
  • 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;267:173–176.
  • Yang C, Shirayama Y, Zhang J-C, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632.
  • Yang C, Han M, Zhang J-C, et al. Loss of parvalbumin-immunoreactivity in mouse brain regions after repeated intermittent administration of esketamine, but not R-ketamine. Psychiatry Res. 2016;239:281–283.
  • Zhang J-C, Li S-X, Hashimoto K. R (-)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacol Biochem Behav. 2014;116:137–141.
  • Zanos P, Moaddel R, Morris PJ, et al. Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharmacol Rev. 2018;70:621–660.
  • 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.
  • Zhang K, Hashimoto K. An update on ketamine and its two enantiomers as rapid-acting antidepressants. Expert Rev Neurother. 2019;19:83–92.
  • Chang L, Zhang K, Pu Y, et al. Comparison of anatidepressant and side effects in mice after intranasal administration of (R,S)-ketamine, (R)-ketamine and (S)-ketamine. Pharmacol Biochem Behav. 2019;181:53–59.
  • Mathisen LC, Skjelbred P, Skoglund LA, et al. Effect of ketamine, an NMDA receptor inhibitor, in acute and chronic orofacial pain. Pain. 1995;61:215–220.
  • Hashimoto K. Ketamine’s antidepressant action: beyond NMDA receptor inhibition. Expert Opin Ther Targets. 2016;20:1389–1392.
  • Moghaddam B, Adams B, Verma A, et al. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17:2921–2927.
  • Autry AE, Adachi M, Nosyreva E, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011;475:91–95.
  • Li N, Lee B, Liu R-J, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329:959–964.
  • Abdallah CG, Averill LA, Gueorguieva R, et al. Rapamycin, an immunosuppressant and mTORC1 inhibitor, triples the antidepressant response rate of ketamine at 2 weeks following treatment. A double-blind, placebo-controlled, cross-over, randomized clinical trial. bioRxiv preprint first posted online. 2018 Dec 19. DOI:10.1101/500959.
  • Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533:481–486.
  • Chaki S. Is metabolism of (R)-ketamine essential for the antidepressant effects? Int J Neuropsychopharmacol. 2018;21:154–156.
  • Li L, Vlisides PE. Ketamine: 50 years of modulating the mind. Front Hum Neurosci. 2016;10:612.
  • Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19:370–380.
  • Williams NR, Heifets BD, Blasey C, et al. Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am J Psychiatry. 2018;175:1205–1215.
  • Zhang K, Hashimoto K. Lack of opioid system in the antidepressant actions of ketamine. Biol Psychiatry. 2019;85:e25–e27.
  • Yang Y, Cui Y, Sang K, et al. Ketamine blocks bursting in the lateral habenula to rapidly relieve depression. Nature. 2018;554:317–322.
  • 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;24:37–43.
  • 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;29:317–324.
  • 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;71:441–447.
  • Brunton L, Hilal-Dandan R, Knollman B Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 13e. Minion Pro by Cenveo® Publisher Services; 2018.
  • Portmann S, Kwan HY, Theurillat R, et al. Enantioselective capillary electrophoresis for identification and characterization of human cytochrome P450 enzymes which metabolize ketamine and norketamine in vitro. J Chromatogr A. 2010;1217:7942–7948.
  • Ihmsen H, Geisslinger G, Schüttler J. Stereoselective pharmacokinetics of ketamine: R(-)-ketamine inhibits the elimination of S(+)-ketamine. Clin Pharmacol Ther. 2001;70:431–438.
  • Janssen Pharmaceuticals inc. SpravatoTM (esketamine hydrochloride) [Package insert]. Copyright Janssen Pharmaceuticals; Titusville, NJ; 2019. Available from: http://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/SPRAVATO-pi.pdf
  • 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;75:139–148.
  • 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;175:620–630.
  • Fedgchin M, Trivedi M, Daly EJ, et al. Randomized, double-blind study of fixed-dosed intranasal esketamine plus oral antidepressant vs. active control in treatment-resistant depression. Presented at the 9th Biennial Conference of the International Society for Affective Disorders (ISAD) and the Houston Mood Disorders Conference. Houston, TX, USA, 2018 September 21.
  • Lam RW, Michalak E, Swinson R. Assessment scales in depression, mania and anxiety. London: Taylor and Francis; 2005.
  • Zimmerman M, Posternak MA, Chelminski I. Derivation of a definition of remission on the Montgomery-Asberg depression rating scale corresponding to the definition of remission on the Hamilton rating scale for depression. J Psychiatr Res. 2004;38:577–582.
  • 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;176:428–438.
  • Ochs-Ross R, Daly EJ, Zhang Y, et al. Efficacy and safety of intranasal esketamine plus an oral antidepressant in elderly patients with treatment-resistant depression. Biol Psychiatry. 2018;83:S391.
  • Joyce PR, Mulder RT, Luty SE, et al. Patterns and predictors of remission, response and recovery in major depression treated with fluoxetine or nortriptyline. Aust N Z J Psychiatry. 2002;36:384–391.
  • Mynors-Wallis L, Gath D. Predictors of treatment outcome for major depression in primary care. Psychol Med. 1997;27:731–736.
  • Daly E, Trivedi M, Janik A, et al. A randomized withdrawal, double-blind, multicenter study of esketamine nasal spray plus an oral antidepressant for relapse prevention in treatment-resistant depression. Poster/abstract presented at Annual Meeting of the American Society of Clinical Psychopharmacology, Hollywood, Florida; May 30, 2018. Poster W68.
  • Wajs E, Aluisio L, Morrison R, et al. Long-term safety of esketamine nasal spray plus an oral antidepressant in patients with treatment-resistant depression: phase 3, open label safety and efficacy study (SUSTAIN-2). Poster presented at: The American Society of Clinical Psychopharmacology Meeting; May 2018; Miami, Florida. 2018.
  • Correia-Melo FS, Leal GC, Carvalho MS, et al. Comparative study of esketamine and racemic ketamine in treatment-resistant depression: protocol for a non-inferiority clinical trial. Medicine (Baltimore). 2018;97:e12414.
  • Short B, Fong J, Galvez V, et al. Side-effects associated with ketamine use in depression: a systematic review. Lancet Psychiatry. 2018;5:65–78.
  • Schatzberg AF. A word to the wise about intranasal esketamine. Am J Psychiatry. 2019;176:422–424.
  • Wan L-B, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76:247–252.
  • FDA report on esketamine for treatment resistant depression. 2019; Accessed April 9, 2019. Available from: https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/PsychopharmacologicDrugsAdvisoryCommittee/UCM630970.pdf.
  • 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;234:3175–3183.
  • Chu PS-K, Ma W-K, Wong SC-W, et al. The destruction of the lower urinary tract by ketamine abuse: a new syndrome?. BJU Int. 2008(102):1616-1622.
  • Shahani R, Streutker C, Dickson B, et al. Ketamine-associated ulcerative cystitis: A new clinical entity. Urology. 2007;69:810–812.
  • Tam Y-H, Ng C-F, Pang KK-Y, et al. One-stop clinic for ketamine-associated uropathy: report on service delivery model, patients’ characteristics and non-invasive investigations at baseline by a cross-sectional study in a prospective cohort of 318 teenagers and young adults. BJU Int. 2014;114:754–760.
  • Tsai J-H, Tsai K-B, Jang M-Y. Ulcerative cystitis associated with ketamine. Am J Addictions. 2008;17:453.
  • 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;235:1107–1119.
  • Morgan CJA, Riccelli M, Maitland CH, et al. Long-term effects of ketamine: evidence for a persisting impairment of source memory in recreational users. Drug Alcohol Depend. 2004;75:301–308.
  • Shiroma PR, Albott CS, Johns B, et al. Neurocognitive performance and serial intravenous subanesthetic ketamine in treatment-resistant depression. Int J Neuropsychopharmacoly. 2014;17:1805–1813.
  • Unpublished data: clinical registry number 108104: a single-center, single-dose, double blind, double dummy, placeo controlled, randomized crossover study to evaluate the abuse potential of intranasal esketamine compared to racemic intravenous ketamine in nondependentm, recreational users of perception-altering drugs.
  • Kolar D. Addictive potential of novel treatments for refractory depression and anxiety. Neuropsychiatr Dis Treat. 2018;14:1513–1519.
  • Nutt D, King LA, Saulsbury W, et al. Development of a rational scale to assess the harm of drugs of potential misuse. Lancet. 2007;369:1047–1053.
  • Royal College of Psychiatrists. Statement on ketamine to treat depression. Position statement cert02/17. Approved by the Royal College of Psychiatrists, Committee on ECT and related treatments: 2017.
  • 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.
  • Yatham LN, Kennedy SH, Parikh SV, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) 2018 guidelines for the management of patients with bipolar disorder. Bipolar Disord. 2018;20(2):97–170.

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.