Skip to main content

Advertisement

Log in

Ketamine in neuropsychiatric disorders: an update

  • Review Article
  • Published:
Neuropsychopharmacology Submit manuscript

Abstract

The discovery of ketamine as a rapid-acting antidepressant led to a new era in the development of neuropsychiatric therapeutics, one characterized by an antidepressant response that occurred within hours or days rather than weeks or months. Considerable clinical research supports the use of—or further research with—subanesthetic-dose ketamine and its (S)-enantiomer esketamine in multiple neuropsychiatric disorders including depression, bipolar disorder, anxiety spectrum disorders, substance use disorders, and eating disorders, as well as for the management of chronic pain. In addition, ketamine often effectively targets symptom domains associated with multiple disorders, such as anxiety, anhedonia, and suicidal ideation. This manuscript: 1) reviews the literature on the pharmacology and hypothesized mechanisms of subanesthetic-dose ketamine in clinical research; 2) describes similarities and differences in the mechanism of action and antidepressant efficacy between racemic ketamine, its (S) and (R) enantiomers, and its hydroxynorketamine (HNK) metabolite; 3) discusses the day-to-day use of ketamine in the clinical setting; 4) provides an overview of ketamine use in other psychiatric disorders and depression-related comorbidities (e.g., suicidal ideation); and 5) provides insights into the mechanisms of ketamine and therapeutic response gleaned from the study of other novel therapeutics and neuroimaging modalities.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Current hypothesized mechanisms of ketamine.

Similar content being viewed by others

References

  1. Mion G. History of anaesthesia: the ketamine story - past, present and future. Eur J Anaesthesiol. 2017;34:571–75.

    Article  PubMed  Google Scholar 

  2. Hirota K, Lambert DG. Ketamine; history and role in anesthetic pharmacology. Neuropharmacology. 2022;216:109171.

    Article  CAS  PubMed  Google Scholar 

  3. Nowak G, Trullas R, Layer RT, Skolnick P, Paul IA. Adaptive changes in the N-methyl-D-aspartate receptor complex after chronic treatment with imipramine and 1-aminocyclopropanecarboxylic acid. J Pharm Exp Ther. 1993;265:1380–6.

    CAS  Google Scholar 

  4. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–4.

    Article  CAS  PubMed  Google Scholar 

  5. Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, et al. A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry. 2010;67:793–802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zarate CA Jr, Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A, et al. Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry. 2012;71:939–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63:856–64.

    Article  CAS  PubMed  Google Scholar 

  8. Monteggia LM, Zarate CAJ. Antidepressant actions of ketamine: from molecular mechanisms to clinical practice. Curr Opin Neurobiol. 2015;30:139–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mkrtchian A, Evans JW, Kraus C, Yuan P, Kadriu B, Nugent AC, et al. Ketamine modulates fronto-striatal circuitry in depressed and healthy individuals. Mol Psychiatry. 2021;26:3292–301.

    Article  CAS  PubMed  Google Scholar 

  10. Kishimoto T, Chawla JM, Hagi K, Zarate CA, Kane JM, Bauer M, 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;46:1459–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sassano-Higgins S, Baron D, Juarez G, Esmaili N, Gold M. A review of ketamine abuse and diversion. Depress Anxiety. 2016;33:718–27.

    Article  CAS  PubMed  Google Scholar 

  12. Li J-H, Vicknasingam B, Cheung Y-W, Zhou W, Wibowo Nurhidayat A, Des, et al. To use or not to use: an update on licit and illicit ketamine use. Subst Abus Rehabil. 2011;2:11–20.

    Article  CAS  Google Scholar 

  13. European Monitoring Centre for Drugs and Drug Addiction (EMCDDA). Luxembourg: Publications Office of the European Union; 2019.

  14. Palamar JJ, Salomone A, Rutherford C, Keyes KM. Extensive underreported exposure to ketamine among electronic dance music party attendees. J Gen Intern Med. 2021;36:235–37.

    Article  PubMed  Google Scholar 

  15. Herd DW, Anderson BJ, Keene NA, Holford NH. Investigating the pharmacodynamics of ketamine in children. Paediatr Anaesth. 2008;18:36–42.

    Article  PubMed  Google Scholar 

  16. MacDonald JF, Miljkovic Z, Pennefather P. Use-dependent block of excitatory amino acid currents in cultured neurons by ketamine. J Neurophysiol. 1987;58:251–66.

    Article  CAS  PubMed  Google Scholar 

  17. Huettner JE, Bean BP. Block of N-methyl-D-aspartate-activated current by the anticonvulsant MK-801: selective binding to open channels. Proc Natl Acad Sci. 1988;85:1307–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Desta Z, Moaddel R, Ogburn ET, Xu C, Ramamoorthy A, Venkata SL, et al. Stereoselective and regiospecific hydroxylation of ketamine and norketamine. Xenobiotica. 2012;42:1076–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rao LK, Flaker AM, Friedel CC, Kharasch ED. Role of cytochrome P4502B6 polymorphisms in ketamine metabolism and clearance. Anesthesiology. 2016;125:1103–12.

    Article  CAS  PubMed  Google Scholar 

  20. Domino EF, Domino SE, Smith RE, Domino LE, Goulet JR, Domino KE, et al. Ketamine kinetics in unmedicated and diazepam-premedicated subjects. Clin Pharm Ther. 1984;36:645–53.

    Article  CAS  Google Scholar 

  21. Schüttler J, Stanski DR, White PF, Trevor A, Horai Y, Verotta D, et al. Pharmacodynamic modeling of the EEG effects of ketamine and its enantiomers in man. J Pharmacokinet Biopharm. 1987;15:241–53.

    Article  PubMed  Google Scholar 

  22. Geisslinger G, Menzel-Soglowek S, Kamp HD, Brune K. Stereoselective high-performance liquid chromatographic determination of the enantiomers of ketamine and norketamine in plasma. J Chromatogr. 1991;568:165–76.

    Article  CAS  PubMed  Google Scholar 

  23. Weiss M, Siegmund W. Pharmacokinetic modeling of ketamine enantiomers and their metabolites after administration of prolonged‐release ketamine with emphasis on 2, 6‐hydroxynorketamines. Clin Pharm Drug Dev. 2022;11:194–206.

    Article  CAS  Google Scholar 

  24. Seeburg PH, Burnashev N, Köhr G, Kuner T, Sprengel R, Monyer H. The NMDA receptor channel: molecular design of a coincidence detector. Recent Prog Horm Res. 1995;50:19–34.

    CAS  PubMed  Google Scholar 

  25. Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010;11:682–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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.

    Article  CAS  PubMed  Google Scholar 

  27. Suzuki K, Monteggia LM. The role of eEF2 kinase in the rapid antidepressnt actions of ketamine. Adv Pharm. 2020;89:79–99.

    Article  CAS  Google Scholar 

  28. Zanos P, Gould TD. Mechanisms of ketamine action as an antidepressant. Mol Psychiatry. 2018;23:801–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yang Y, Cui Y, Sang K, Dong Y, Ni Z, Ma S, et al. Ketamine blocks bursting in the lateral habenula to rapidly relieve depression. Nature. 2018;554:317–22.

    Article  CAS  PubMed  Google Scholar 

  30. Zanos P, Moaddel R, Morris PJ, Riggs LM, Highland JN, Georgiou P, et al. Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharm Rev. 2018;70:621–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lumsden EW, Troppoli TA, Myers SJ, Zanos P, Aracava Y, Kehr J, et al. Antidepressant-relevant concentrations of the ketamine metabolite (2R,6R)-hydroxynorketamine do not block NMDA receptor function. Proc Natl Acad Sci USA. 2019;116:5160–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Matveychuk D, Thomas RK, Swainson J, Khullar A, MacKay M-A, Baker GB, et al. Ketamine as an antidepressant: overview of its mechanisms of action and potential predictive biomarkers. Ther Adv Psychopharmacol. 2020;10:2045125320916657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bahji A, Zarate CAJ. Ketamine for bipolar depression: a systematic review. Int J Neuropsychopharmacol. 2021;24:535–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Marcantoni WS, Akoumba BS, Wassef M, Mayrand J, Lai H, Richard-Devantoy S, et al. A systematic review and meta-analysis of the efficacy of intravenous ketamine infusion for treatment resistant depression: January 2009 - January 2019. J Affect Disord. 2020;277:831–41.

    Article  CAS  PubMed  Google Scholar 

  35. McIntyre RS, Rosenblat JD, Nemeroff CB, Sanacora G, Murrough JW, Berk M, et al. Synthesizing the evidence for ketamine and esketamine in treatment-resistant depression: an international expert opinion on the available evidence and implementation. Am J Psychiatry. 2021;178:383–99.

  36. Bahji A, Vazquez GH. Comparative efficacy of racemic ketamine and esketamine for depression: a systematic review and meta-analysis. J Affect Disord. 2021;278:542–55.

    Article  CAS  PubMed  Google Scholar 

  37. Correia-Melo FS, Leal GC, Vieira F, Jesus-Nunes AP, Mello RP, Magnavita G, et al. Efficacy and safety of adjunctive therapy using esketamine or racemic ketamine for adult treatment-resistant depression: A randomized, double-blind, non-inferiority study. J Affect Disord. 2020;264:527–34.

    Article  CAS  PubMed  Google Scholar 

  38. Singh B, Kung S, Pazdernik V, Schak KM, Geske J, Schulte PJ, et al. Comparative effectiveness of intravenous ketamine and intranasal esketamine in clinical practice among patients with treatment-refractory depression: an observational study. J Clin Psychiatry. 2023;84:22m14548.

    Article  PubMed  Google Scholar 

  39. Williamson DJ, Gogate JP, Kern Sliwa JK, Manera LS, Preskorn SH, Winokur A, et al. Longitudinal course of adverse events with esketamine nasal spray: a post hoc analysis of pooled data from Phase 3 trials in patients with treatment-resistant depression. J Clin Psychiatry. 2022;83:21m14318.

    Article  PubMed  Google Scholar 

  40. Fukumoto K, Toki H, Iijima M, Hashihayata T, Yamaguchi J-I, Hashimoto K, et al. Antidepressant potential of (R)-ketamine in rodent models: comparison with (S)-ketamine. J Pharm Exp Ther. 2017;361:9–16.

    Article  CAS  Google Scholar 

  41. Kamp J, van Velzen M, Aarts L, Niesters M, Dahan A, Olofsen E. Stereoselective ketamine effect on cardiac output: a population pharmacokinetic/pharmacodynamic modelling study in healthy volunteers. Br J Anaesth. 2021;127:23–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Leal GC, Bandeira ID, Correia-Melo FS, Telles M, Mello RP, Vieira F, et al. Intravenous arketamine for treatment-resistant depression: open-label pilot study. Eur Arch Psychiatr Clin Neurosci. 2021;271:577–82.

    Article  Google Scholar 

  43. Atai Life Sciences. atai Life Sciences announces results from Phase 2a trial of PCN-101 (R-ketamine) for treatment-resistant depression. 2023. https://www.globenewswire.com/news-release/2023/01/06/2584334/0/en/atai-Life-Sciences-Announces-Results-from-Phase-2a-Trial-of-PCN-101-R-ketamine-for-Treatment-Resistant-Depression.html.

  44. Leal GC, Souza-Marques B, Mello RP, Bandeira ID, Caliman-Fontes AT, Carneiro BA, et al. Arketamine as adjunctive therapy for treatment-resistant depression: a placebo-controlled pilot study. J Affect Disord. 2023;330:7–15.

    Article  CAS  PubMed  Google Scholar 

  45. Zanos P, Highland JN, Liu X, Troppoli TA, Georgiou P, Lovett J, et al. (R)-Ketamine exerts antidepressant actions partly via conversion to (2R,6R)-hydroxynorketamine, while causing adverse effects at sub-anaesthetic doses. Br J Pharm. 2019;176:2573–92.

    Article  CAS  Google Scholar 

  46. Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533:481–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Grunebaum MF, Galfalvy HC, Choo TH, Parris MS, Burke AK, Suckow RF, et al. Ketamine metabolite pilot study in a suicidal depression trial. J Psychiatr Res. 2019;117:129–34.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Hashimoto K, Yang C. Is (S)-norketamine an alternative antidepressant for esketamine? Eur Arch Psychiatry Clin Neurosci. 2019;269:867–68.

    Article  PubMed  Google Scholar 

  49. Zheng W, Li X-H, Zhu X-M, Cai D-B, Yang X-H, Ungvari GS, et al. Adjunctive ketamine and electroconvulsive therapy for major depressive disorder: a meta-analysis of randomized controlled trials. J Affect Disord. 2019;250:123–31.

    Article  CAS  PubMed  Google Scholar 

  50. Dong J, Min S, Qiu H, Chen Q, Ren L. Intermittent administration of low dose ketamine can shorten the course of electroconvulsive therapy for depression and reduce complications: a randomized controlled trial. Psychiatry Res. 2019;281:112573.

    Article  PubMed  Google Scholar 

  51. Zou W, Min S, Chen Q, Li X, Ren L. Subanesthetic dose of ketamine for the antidepressant effects and the associated cognitive impairments of electroconvulsive therapy in elderly patients—a randomized, double‐blind, controlled clinical study. Brain Behav. 2021;11:e01775.

    Article  CAS  PubMed  Google Scholar 

  52. Li D-J, Wang F-C, Chu C-S, Chen T-Y, Tang C-H, Yang W-C, et al. Significant treatment effect of add-on ketamine anesthesia in electroconvulsive therapy in depressive patients: a meta-analysis. Eur Neuropsychopharmacol. 2017;27:29–41.

    Article  CAS  PubMed  Google Scholar 

  53. McGirr A, Berlim MT, Bond DJ, Neufeld NH, Chan PY, Yatham LN, et al. A systematic review and meta-analysis of randomized controlled trials of adjunctive ketamine in electroconvulsive therapy: efficacy and tolerability. J Psychiatr Res. 2015;62:23–30.

    Article  PubMed  Google Scholar 

  54. Di Vincenzo JD, Lipsitz O, Rodrigues NB, Lee Y, Gill H, Kratiuk K, et al. Ketamine monotherapy versus adjunctive ketamine in adults with treatment-resistant depression: results from the Canadian Rapid Treatment Centre of Excellence. J Psychiatr Res. 2021;143:209–14.

    Article  PubMed  Google Scholar 

  55. Zheng W, Zhou Y-L, Liu W-J, Wang C-Y, Zhan Y-N, Lan X-F, et al. A preliminary study of adjunctive ketamine for treatment-resistant bipolar depression. J Affect Disord. 2020;275:38–43.

    Article  CAS  PubMed  Google Scholar 

  56. Price RB, Spotts C, Panny B, Griffo A, Degutis M, Cruz N, et al. A novel, brief, fully automated intervention to extend the antidepressant effect of a single ketamine infusion: a randomized clinical trial. Am J Psychiatry. 2022;179:959–68.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Wilkinson ST, Rhee TG, Joorman J, Webler R, Lopez MO, Kitay B, et al. Cognitive behavioral therapy to sustain the antidepressant effects of ketamine in treatment-resistant depression: a randomized clinical trial. Psychother Psychosom. 2021;90:318–27.

    Article  PubMed  Google Scholar 

  58. McInnes LA, Qian JJ, Gargeya RS, DeBattista C, Heifets BD. A retrospective analysis of ketamine intravenous therapy for depression in real-world care settings. J Affect Disord. 2022;301:486–95.

    Article  CAS  PubMed  Google Scholar 

  59. Alnefeesi Y, Chen-Li D, Krane E, Jawad MY, Rodgrigues NB, Ceban F, et al. Real-world effectiveness of ketamine in treatment-resistant depression: a systematic review & meta-analysis. J Psychiatr Res. 2022;151:693–709.

    Article  PubMed  Google Scholar 

  60. Lipsitz O, Di Vincenzo JD, Rodrigues NB, Cha DS, Lee Y, Greenberg D, et al. Safety, tolerability, and real-world effectiveness of intravenous ketamine in older adults with treatment-resistant depression: a case series. Am J Geriatr Psychiatry. 2021;29:899–913.

    Article  PubMed  Google Scholar 

  61. Fancy F, Rodrigues NB, Di Vincenzo JD, Chau EH, Sethi R, Husain MI, et al. Real world effectiveness of repeated ketamine infusions for treatment resistant bipolar depression. Bipolar Disord. 2023;25:99–109.

  62. Daly EJ, Trivedi MH, Janik A, Li H, Zhang Y, Li X, et al. Efficacy of esketamine nasal spray plus oral antidepressant treatment for relapse prevention in patients with treatment-resistant depression: a randomized clinical trial. JAMA Psychiatry. 2019;76:893–903.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Fu D-J, Ionescu DF, Li X, Lane R, Lim P, Sanacora G, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81:19m13191.

    Article  PubMed  Google Scholar 

  64. Ionescu DF, Fu D-J, Qiu X, Lane R, Lim P, Kasper S, et al. Esketamine nasal spray for rapid reduction of depressive symptoms in patients with major depressive disorder who have active suicide ideation with intent: results of a Phase 3, double-blind, randomized study (ASPIRE II). Int J Neuropsychopharmacol. 2021;24:22–31.

    Article  PubMed  Google Scholar 

  65. Popova V, Daly EJ, Trivedi M, Cooper K, Lane R, Lim P, 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;176:428–38.

    Article  PubMed  Google Scholar 

  66. Hull TD, Malgaroli M, Gazzaley A, Akiki TJ, Madan A, Vando L, et al. At-home, sublingual ketamine telehealth is a safe and effective treatment for moderate to severe anxiety and depression: findings from a large, prospective, open-label effectiveness trial. J Affect Disord. 2022;314:59–67.

    Article  CAS  PubMed  Google Scholar 

  67. Samalin L, Rothärmel M, Mekaoui L, Gaudré-Wattinne E, Codet M-A, Bouju S, et al. Esketamine nasal spray in patients with treatment-resistant depression: the real-world experience in the French cohort early-access programme. Int J Psychiatry Clin Pr. 2022;26:352–62.

    Article  CAS  Google Scholar 

  68. Simmler LD, Li Y, Hadjas LC, Hiver A, van Zessen R, Lüscher C. Dual action of ketamine confines addiction liability. Nature. 2022;608:368–73.

    Article  CAS  PubMed  Google Scholar 

  69. Eckstrand J, Fattah C, Persson M, Cheng T, Nordanskog P, Åkeson J, et al. Racemic ketamine as an alternative to electroconvulsive therapy for unipolar depression. A randomized, open-label, non-inferiority trial (KetECT). Int J Neuropsychopharmacol. 2022;25:339–49.

    Article  Google Scholar 

  70. Rhee TG, Shim SR, Forester BP, Nierenberg AA, McIntyre RS, Papakostas GI, et al. Efficacy and safety of ketamine vs electroconvulsive therapy among patients with major depressive episode: a systematic review and meta-analysis. JAMA Psychiatry. 2022;79:1162–72.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Papadimitropoulou K, Vossen C, Karabis A, Donatti C, Kubitz N. 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;33:701–11.

    Article  CAS  PubMed  Google Scholar 

  72. Wilkinson ST, Ballard ED, Bloch MH, Mathew SJ, Murrough JW, Feder A, et al. The effect of a single dose of intravenous ketamine on suicidal ideation: a systematic review and individual participant data meta-analysis. Am J Psychiatry. 2018;175:150–58.

    Article  PubMed  Google Scholar 

  73. Hochschild A, Grunebaum MF, Mann JJ. The rapid anti-suicidal ideation effect of ketamine: a systematic review. Prev Med. 2021;152:106524.

    Article  PubMed  Google Scholar 

  74. Witt K, Potts J, Hubers A, Grunebaum MF, Murrough JW, Loo C, et al. Ketamine for suicidal ideation in adults with psychiatric disorders: a systematic review and meta-analysis of treatment trials. Aust N Z J Psychiatry. 2020;54:29–45.

    Article  PubMed  Google Scholar 

  75. Alario AA, Niciu MJ. (Es)Ketamine for suicidal ideation and behavior: clinical efficacy. Chronic Stress. 2022;6:24705470221128017.

  76. Abbar M, Demattei C, El-Hage W, Llorca P-M, Samalin L, Demaricourt P, et al. Ketamine for the acute treatment of severe suicidal ideation: double blind, randomised placebo controlled trial. BMJ. 2022;376:e067194.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Domany Y, Shelton RC, mcCullumsmith CB. Ketamine for acute suicidal ideation. An emergency department intervention: a randomized, double‐blind, placebo‐controlled, proof‐of‐concept trial. Depress Anxiety. 2020;37:224–33.

    Article  CAS  PubMed  Google Scholar 

  78. Shivanekar S, Gopalan P, Pizon A, Spotts C, Cruz N, Lightfoot M, et al. A pilot study of ketamine infusion after suicide attempt: new frontiers in treating acute suicidality in a real-world medical setting. Int J Environ Res Public Health. 2022;19:13792.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Surjan J, Grossi JD, Del Porto JA, Delfino RS, de Oliveira Cerqueira R, Lucchese AC, et al. Efficacy and safety of subcutaneous esketamine in the treatment of suicidality in major depressive disorder and bipolar depression. Clin Drug Investig. 2022;42:865–73.

    Article  CAS  PubMed  Google Scholar 

  80. Zhan Y, Zhang B, Zhou Y, Zheng W, Liu W, Wang C, et al. A preliminary study of anti-suicidal efficacy of repeated ketamine infusions in depression with suicidal ideation. J Affect Disord. 2019;251:205–12.

    Article  CAS  PubMed  Google Scholar 

  81. Zhou Y, Wang C, Lan X, Zheng W, Li H, Chao Z, et al. The effectiveness of repeated intravenous ketamine on subjective and objective psychosocial function in patients with treatment-resistant depression and suicidal ideation. J Affect Disord. 2022;304:78–84.

    Article  CAS  PubMed  Google Scholar 

  82. Oliver PA, Snyder AD, Feinn R, Malov S, McDiarmid G, Arias AJ. Clinical effectiveness of intravenous racemic ketamine infusions in a large community sample of patients with treatment-resistant depression, suicidal ideation, and generalized anxiety symptoms: a retrospective chart review. J Clin Psychiatry. 2022;83:21m14336.

    Article  PubMed  Google Scholar 

  83. Phillips JL, Norris S, Talbot J, Hatchard T, Ortiz A, Birmingham M, et al. Single and repeated ketamine infusions for reduction of suicidal ideation in treatment-resistant depression. Neuropsychopharmacology. 2020;45:606–12.

    Article  CAS  PubMed  Google Scholar 

  84. Chen X, Wang M, Hu Y, Zhan Y, Zhou Y, Zheng W, et al. Working memory associated with anti-suicidal ideation effect of repeated-dose intravenous ketamine in depressed patients. Eur Arch Psychiatr Clin Neurosci. 2021;271:431–38.

    Article  Google Scholar 

  85. Zhou Y, Liu W, Zheng W, Wang C, Zhan Y, Lan X, et al. Predictors of response to repeated ketamine infusions in depression with suicidal ideation: an ROC curve analysis. J Affect Disord. 2020;264:263–71.

    Article  CAS  PubMed  Google Scholar 

  86. McIntyre RS, Rodrigues NB, Lipsitz O, Nasri F, Gill H, Lui LM, et al. The effectiveness of intravenous ketamine in adults with treatment-resistant major depressive disorder and bipolar disorder presenting with prominent anxiety: results from the Canadian Rapid Treatment Center of Excellence. J Psychopharmacol. 2021;35:128–36.

    Article  PubMed  Google Scholar 

  87. Ionescu DF, Bentley KH, Eikermann M, Taylor N, Johnson-Akeju O, Swee MB, et al. Repeat-dose ketamine augmentation for treatment-resistant depression with chronic suicidal ideation: a randomized, double blind, placebo controlled trial. J Affect Disord. 2019;243:516–24.

    Article  CAS  PubMed  Google Scholar 

  88. Fedgchin M, Trivedi M, Daly EJ, Melkote R, Lane R, Lim P, et al. Efficacy and safety of fixed-dose esketamine nasal spray combined with a new oral antidepressant in treatment-resistant depression: results of a randomized, double-blind, active-controlled study (TRANSFORM-1). Int J Neuropsychopharmacol. 2019;22:616–30.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Ochs-Ross R, Daly EJ, Zhang Y, Lane R, Lim P, Morrison RL, et al. Efficacy and safety of esketamine nasal spray plus an oral antidepressant in elderly patients with treatment-resistant depression—TRANSFORM-3. Am J Geriatr Psychiatry. 2020;28:121–41.

    Article  PubMed  Google Scholar 

  90. Jollant F, Colle R, Nguyen TML, Corruble E, Gardier AM, Walter M, et al. Ketamine and esketamine in suicidal thoughts and behaviors: a systematic review. Ther Adv Psychopharmacol. 2023;13:20451253231151327.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Bloomfield-Clagett B, Ballard ED, Greenstein DK, Wilkinson ST, Grunebaum MF, Murrough JW, et al. A participant-level integrative data analysis of differential placebo response for suicidal ideation and nonsuicidal depressive symptoms in clinical trials of intravenous racemic ketamine. Int J Neuropsychopharmacol. 2022;25:827–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Averill LA, Averill CL, Gueorguieva R, Fouda S, Sherif M, Ahn K-H, et al. mTORC1 inhibitor effects on rapid ketamine-induced reductions in suicidal ideation in patients with treatment-resistant depression. J Affect Disord. 2022;303:91–97.

    Article  CAS  PubMed  Google Scholar 

  93. Abdallah CG, Averill LA, Gueorguieva R, Goktas S, Purohit P, Ranganathan M, et al. Modulation of the antidepressant effects of ketamine by the mTORC1 inhibitor rapamycin. Neuropsychopharmacology. 2020;45:990–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. McIntyre RS, Lipsitz O, Rodrigues NB, Lee Y, Cha DS, Vinberg M, et al. The effectiveness of ketamine on anxiety, irritability, and agitation: implications for treating mixed features in adults with major depressive or bipolar disorder. Bipolar Disord. 2020;22:831–40.

    Article  CAS  PubMed  Google Scholar 

  95. Whittaker E, Dadabayev AR, Joshi SA, Glue P. Systematic review and meta-analysis of randomized controlled trials of ketamine in the treatment of refractory anxiety spectrum disorders. Ther Adv Psychopharmacol. 2021;11:20451253211056743.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Ahuja S, Brendle M, Smart L, Moore C, Thielking P, Robison R. Real-world depression, anxiety and safety outcomes of intramuscular ketamine treatment: a retrospective descriptive cohort study. BMC Psychiatry. 2022;22:634.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Glue P, Medlicott NJ, Neehoff SM, Surman P, Lam F, Hung N, et al. Safety and efficacy of extended release ketamine tablets in patients with treatment-resistant depression and anxiety: open label pilot study. Ther Adv Psychopharmacol. 2020;10:2045125320922474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Taylor JH, Landeros-Weisenberger A, Coughlin C, Mulqueen J, Johnson JA, Gabriel D, et al. Ketamine for social anxiety disorder: a randomized, placebo-controlled crossover trial. Neuropsychopharmacology. 2018;43:325–33.

    Article  CAS  PubMed  Google Scholar 

  99. Glue P, Neehoff SM, Medlicott NJ, Gray A, Kibby G, McNaughton N. Safety and efficacy of maintenance ketamine treatment in patients with treatment-refractory generalised anxiety and social anxiety disorders. J Psychopharmacol. 2018;32:663–67.

    Article  CAS  PubMed  Google Scholar 

  100. Hoskins M, Pearce J, Bethell A, Dankova L, Barbui C, Tol WA, et al. Pharmacotherapy for post-traumatic stress disorder: systematic review and meta-analysis. Br J Psychiatry. 2015;206:93–100.

    Article  PubMed  Google Scholar 

  101. Feder A, Parides MK, Murrough JW, Perez AM, Morgan JE, Saxena S, et al. Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA Psychiatry. 2014;71:681–88.

    Article  CAS  PubMed  Google Scholar 

  102. Feder A, Costi S, Rutter SB, Collins AB, Govindarajulu U, Jha MK, et al. A randomized controlled trial of repeated ketamine administration for chronic posttraumatic stress disorder. Am J Psychiatry. 2021;178:193–202.

    Article  PubMed  Google Scholar 

  103. Ross C, Jain R, Bonnett CJ, Wolfson P. High-dose ketamine infusion for the treatment of posttraumatic stress disorder in combat veterans. Ann Clin Psychiatry. 2019;31:271–79.

    PubMed  Google Scholar 

  104. Abdallah CG, Roache JD, Gueorguieva R, Averill LA, Young-McCaughan S, Shiroma PR, et al. Dose-related effects of ketamine for antidepressant-resistant symptoms of posttraumatic stress disorder in veterans and active duty military: a double-blind, randomized, placebo-controlled multi-center clinical trial. Neuropsychopharmacology. 2022;47:1574–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Davis MT, DellaGioia N, Maruff P, Pietrzak RH, Esterlis I. Acute cognitive effects of single-dose intravenous ketamine in major depressive and posttraumatic stress disorder. Transl Psychiatry. 2021;11:205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Al Jumaili W, Trivedi C, Chao T, Kubosumi A, Jain S. The safety and efficacy of ketamine NMDA receptor blocker as a therapeutic intervention for PTSD review of a randomized clinical trial. Behav Brain Res. 2022;424:113804.

    Article  Google Scholar 

  107. Dadabayev AR, Joshi SA, Reda MH, Lake T, Hausman MS, Domino E, et al. Low dose ketamine infusion for comorbid posttraumatic stress disorder and chronic pain: a randomized double-blind clinical trial. Chronic Stress. 2020;4:2470547020981670.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Rodriguez CI, Kegeles LS, Levinson A, Feng T, Marcus SM, Vermes D, et al. Randomized controlled crossover trial of ketamine in obsessive-compulsive disorder: proof-of-concept. Neuropsychopharmacology. 2013;38:2475–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Rodriguez CI, Kegeles LS, Flood P, Simpson HB. Rapid resolution of obsessions after an infusion of intravenous ketamine in a patient with treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2011;72:16895.

    Article  Google Scholar 

  110. Bottemanne H, Arnould A. Ketamine augmentation of exposure response prevention therapy for obsessive-compulsive disorder. Innov Clin Neurosci. 2021;18:9–11.

    PubMed  PubMed Central  Google Scholar 

  111. Milad MR, Furtak SC, Greenberg JL, Keshaviah A, Im JJ, Falkenstein MJ, et al. Deficits in conditioned fear extinction in obsessive-compulsive disorder and neurobiological changes in the fear circuit. JAMA Psychiatry. 2013;70:608–18.

    Article  PubMed  Google Scholar 

  112. Sharma LP, Thamby A, Balachander S, Janardhanan CN, Jaisoorya TS, Arumugham SS, et al. Clinical utility of repeated intravenous ketamine treatment for resistant obsessive-compulsive disorder. Asian J Psychiatr. 2020;52:102183.

    Article  PubMed  Google Scholar 

  113. van Roessel P, Asgari S, Jo B, Williams L, Rodriguez C. P415. Cognitive control predicts alleviation of OCD symptoms by ketamine. ACNP 61st Annu Meet Poster Abstr Neuropsychopharmacol 2022;47:302.

    Google Scholar 

  114. Rodriguez C, Chen C-M, Glover G, Jo B, Spielman D, Williams L, et al. P416. Efficacy of ketamine in unmedicated adults with obsessive-compulsive disorder: a randomized controlled trial. ACNP 61st Annu Meet Poster Abstr Neuropsychopharmacol 2022;47:302–03.

    Google Scholar 

  115. Bloch MH, Wasylink S, Landeros-Weisenberger A, Panza KE, Billingslea E, Leckman JF, et al. Effects of ketamine in treatment-refractory obsessive-compulsive disorder. Biol Psychiatry. 2012;72:964–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Niciu MJ, Grunschel BDG, Corlett PR, Pittenger C, Bloch MH. Two cases of delayed-onset suicidal ideation, dysphoria and anxiety after ketamine infusion in patients with obsessive-compulsive disorder and a history of major depressive disorder. J Psychopharmacol. 2013;27:651–54.

    Article  PubMed  Google Scholar 

  117. Bandeira ID, Lins-Silva DH, Cavenaghi VB, Dorea-Bandeira I, Faria-Guimarães D, Barouh JL, et al. Ketamine in the treatment of obsessive-compulsive disorder: a systematic review. Harv Rev Psychiatry. 2022;30:135–45.

    Article  PubMed  Google Scholar 

  118. Schmidt U, Adan R, Böhm I, Campbell IC, Dingemans A, Ehrlich S, et al. Eating disorders: the big issue. Lancet Psychiatry. 2016;3:313–5.

    Article  PubMed  Google Scholar 

  119. Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. Arch Gen Psychiatry. 2011;68:724–31.

    Article  PubMed  Google Scholar 

  120. Wild B, Friederich HC, Zipfel S, Resmark G, Giel K, Teufel M, et al. Predictors of outcomes in outpatients with anorexia nervosa - Results from the ANTOP study. Psychiatry Res. 2016;244:45–50.

    Article  PubMed  Google Scholar 

  121. Calabrese L, Scolnick B, Zupec-Kania B, Beckwith C, Costello K, Frank GKW. Ketogenic diet and ketamine infusion treatment to target chronic persistent eating disorder psychopathology in anorexia nervosa: a pilot study. Eat Weight Disord. 2022;27:3751–57.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Robison R, Lafrance A, Brendle M, Smith M, Moore C, Ahuja S, et al. A case series of group-based ketamine-assisted psychotherapy for patients in residential treatment for eating disorders with comorbid depression and anxiety disorders. J Eat Disord. 2022;10:65.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Schwartz T, Trunko ME, Feifel D, Lopez E, Peterson D, Frank GKW, et al. A longitudinal case series of IM ketamine for patients with severe and enduring eating disorders and comorbid treatment-resistant depression. Clin Case Rep. 2021;9:e03869.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Hedges DW, Reimherr FW, Hoopes SP, Rosenthal NR, Kamin M, Capece JA. Treatment of bulimia nervosa with topiramate in a randomized, double-blind, placebo-controlled trial, part 2: improvement in psychiatric measures. J Clin Psychiatry. 2003;64:21395.

    Article  Google Scholar 

  125. Hoopes SP, Reimherr FW, Hedges DW, Rosenthal NR, Kamin M, Karim R, et al. Treatment of bulimia nervosa with topiramate in a randomized, double-blind, placebo-controlled trial, part 1: improvement in binge and purge measures. J Clin Psychiatry. 2003;64:1335–41.

    Article  CAS  PubMed  Google Scholar 

  126. Nickel C, Tritt K, Muehlbacher M, Pedrosa Gil F, Mitterlehner FO, Kaplan P, et al. Topiramate treatment in bulimia nervosa patients: a randomized, double-blind, placebo-controlled trial. Int J Eat Disord. 2005;38:295–300.

    Article  PubMed  Google Scholar 

  127. Haas DA, Harper DG. Ketamine: a review of its pharmacologic properties and use in ambulatory anesthesia. Anesth Prog. 1992;39:61–68.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Collins S, Sigtermans MJ, Dahan A, Zuurmond WWA, Perez RSGM. NMDA receptor antagonists for the treatment of neuropathic pain. Pain Med Malden Mass. 2010;11:1726–42.

    Google Scholar 

  129. Orhurhu V, Orhurhu MS, Bhatia A, Cohen SP. Ketamine infusions for chronic pain: a systematic review and meta-analysis of randomized controlled trials. Anesth Analg. 2019;129:241–54.

    Article  CAS  PubMed  Google Scholar 

  130. Lumanauw DD, Youn S, Horeczko T, Yadav K, Tanen DA. Subdissociative‐dose ketamine is effective for treating acute exacerbations of chronic pain. Acad Emerg Med. 2019;26:1044–51.

    Article  PubMed  Google Scholar 

  131. Pickering G, Pereira B, Morel V, Corriger A, Giron F, Marcaillou F, et al. Ketamine and magnesium for refractory neuropathic pain: A randomized, double-blind, crossover trial. Anesthesiology. 2020;133:154–64.

    Article  CAS  PubMed  Google Scholar 

  132. Corriger A, Voute M, Lambert C, Pereira B, Pickering G. OKAPI Consortium. Ketamine for refractory chronic pain: a 1-year follow-up study. Pain. 2022;163:690–701.

    Article  CAS  PubMed  Google Scholar 

  133. Motoyama Y, Oshiro Y, Takao Y, Sato H, Obata N, Izuta S, et al. Resting-state brain functional connectivity in patients with chronic pain who responded to subanesthetic-dose ketamine. Sci Rep. 2019;9:12912.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Schulman EA, Lake AER, Goadsby PJ, Peterlin BL, Siegel SE, Markley HG, et al. Defining refractory migraine and refractory chronic migraine: proposed criteria from the Refractory Headache Special Interest Section of the American Headache Society. Headache. 2008;48:778–82.

    Article  PubMed  Google Scholar 

  135. Schwenk ES, Torjman MC, Moaddel R, Lovett J, Katz D, Denk W, et al. Ketamine for refractory chronic migraine: an observational pilot study and metabolite analysis. J Clin Pharm. 2021;61:1421–29.

    Article  CAS  Google Scholar 

  136. Yuan H, Natekar A, Park JS, Lauritsen C, Viscusi ER, Marmura MJ. Real-world study of intranasal ketamine for use in patients with refractory chronic migraine. medRxiv. 2022. https://www.medrxiv.org/content/10.1101/2022.11.20.22282558v1.

  137. Turner AL, Shandley S, Miller E, Perry MS, Ryals B. Intranasal ketamine for abortive migraine therapy in pediatric patients: a single-center review. Pediatr Neurol. 2020;104:46–53.

    Article  PubMed  Google Scholar 

  138. Benish T, Villalobos D, Love S, Casmaer M, Hunter CJ, Summers SM, et al. The THINK (Treatment of Headache with Intranasal Ketamine) trial: a randomized controlled trial comparing intranasal ketamine with intravenous metoclopramide. J Emerg Med. 2019;56:248–47.

    Article  PubMed  Google Scholar 

  139. Chah N, Jones M, MIlord S, Al-Eryani K, Enciso R. Efficacy of ketamine in the treatment of migraines and other unspecified primary headache disorders compared to placebo and other interventions: a systematic review. J Dent Anesth Pain Med. 2021;21:413–29.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Nagy J. Alcohol related changes in regulation of NMDA receptor functions. Curr Neuropharmacol. 2008;6:39–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Das RK, Gale G, Walsh K, Hennessy VE, Iskandar G, Mordecai LA, et al. Ketamine can reduce harmful drinking by pharmacologically rewriting drinking memories. Nat Commun. 2019;10:5187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Grabski M, McAndrew A, Lawn W, Marsh B, Raymen L, Stevens T, et al. Adjunctive ketamine with relapse prevention-based psychological therapy in the treatment of alcohol use disorder. Am J Psychiatry. 2022;179:152–62.

  143. Dakwar E, Levin F, Hart CL, Basaraba C, Choi J, Pavlicova M, et al. A single ketamine infusion combined with motivational enhancement therapy for alcohol use disorder: a randomized midazolam-controlled pilot trial. Am J Psychiatry. 2020;177:125–33.

    Article  PubMed  Google Scholar 

  144. Mollaahmetoglu OM, Keeler J, Ashbullby KJ, Ketzitzidou-Argyri E, Grabski M, Morgan CJA. This is something that changed my life”: a qualitative study of patients’ experiences in a clinical trial of ketamine treatment for alcohol use disorders. Front Psychiatry. 2021;12:695335.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Garel N, McAnulty C, Greenway KT, Lesperance P, Miron J-P, Rej S, et al. Efficacy of ketamine intervention to decrease alcohol use, cravings, and withdrawal symptoms in adults with problematic alcohol use or alcohol use disorder: a systematic review and comprehensive analysis of mechanism of actions. Drug Alcohol Depend. 2022;239:109606.

    Article  CAS  PubMed  Google Scholar 

  146. Azhari N, Hu H, O’Malley KY, Blocker ME, Levin FR, Dakwar E. Ketamine-facilitated behavioral treatment for cannabis use disorder: a proof of concept study. Am J Drug Alcohol Abus. 2021;47:92–97.

    Article  Google Scholar 

  147. Dakwar E, Levin F, Foltin RW, Nunes EV, Hart CL. The effects of subanesthetic ketamine infusions on motivation to quit and cue-induced craving in cocaine-dependent research volunteers. Biol Psychiatry. 2014;76:40–46.

    Article  CAS  PubMed  Google Scholar 

  148. Dakwar E, Hart CL, Levin F, Nunes EV, Foltin RW. Cocaine self-administration disrupted by the N-methyl-D-aspartate receptor antagonist ketamine: a randomized, crossover trial. Mol Psychiatry. 2017;22:76–81.

    Article  CAS  PubMed  Google Scholar 

  149. Dakwar E, Nunes EV, Hart CL, Foltin RW, Mathew SJ, Carpenter KM, et al. A single ketamine infusion combined with mindfulness-based behavioral modification to treat cocaine dependence: a randomized clinical trial. Am J Psychiatry. 2019;176:923–30.

    Article  PubMed  Google Scholar 

  150. Hailozian C, Luftig J, Liang A, Outhay M, Ullal M, Anderson ES, et al. Synergistic effect of ketamine and buprenorphine observed in the treatment of buprenorphine precipitated opioid withdrawal in a patient with fentanyl use. J Addict Med. 2022;16:483–87.

    Article  CAS  PubMed  Google Scholar 

  151. Anghelescu DL, Ryan S, Wu D, Morgan KJ, Patni T, Li Y. Low‐dose ketamine infusions reduce opioid use in pediatric and young adult oncology patients. Pediatr Blood Cancer. 2022;69:e29693.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Boenigk K, Echevarria GC, Nisimov E, von Bergen Granell AE, Cuff GE, Wang J, et al. Low-dose ketamine infusion reduces postoperative hydromorphone requirements in opioid-tolerant patients following spinal fusion: a randomised controlled trial. Eur J Anesthesiol. 2019;36:8–15.

    Article  CAS  Google Scholar 

  153. Sahmeddini MA, Khosravi MB, Farbood A. Comparison of perioperative systemic lidocaine or systemic ketamine in acute pain management of patients with opioid use disorder after orthopedic surgery. J Addict Med. 2019;13:220–26.

    Article  CAS  PubMed  Google Scholar 

  154. Le TT, Cordero IP, Jawad MY, Swainson J, Di Vincenzo JD, Jaberi S, et al. The abuse liability of ketamine: a scoping review of preclinical and clinical studies. J Psychiatr Res. 2022;151:476–96.

    Article  PubMed  Google Scholar 

  155. Bonaventura J, Lam S, Carlton M, Boehm MA, Gomez JL, Solis O, et al. Pharmacological and behavioral divergence of ketamine enantiomers: implications for abuse liability. Mol Psychiatry. 2021;26:6704–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Chang LC, Zhang K, Pu Y, Qu Y, Wang S-M, Xiong Z, et al. Comparison of antidepressant and side effects in mice after intranasal administration of (R,S)-ketamine, (R)-ketamine, and (S)-ketamine. Pharm Biochem Behav. 2019;181:53–59.

    Article  CAS  Google Scholar 

  157. Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Acevedo-Diaz EE, Cavanaugh GW, Greenstein D, Kraus C, Kadriu B, Zarate CAJ, et al. Comprehensive assessment of side effects associated with a single dose of ketamine in treatment-resistant depression. J Affect Disord. 2020;263:568–75.

    Article  CAS  PubMed  Google Scholar 

  159. Feifel D, Dadiomov D, Lee KC. Safety of repeated administration of parenteral ketamine for depression. Pharmaceuticals. 2020;13:151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Wan LB, Levitch CF, Perez AM, Brallier JW, Iosifescu DV, Chang LC, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76:247–52.

    Article  PubMed  Google Scholar 

  161. Enck P, Bingel U, Schedlowski M, Rief W. The placebo response in medicine: minimize, maximize or personalize? Nat Rev Drug Discov. 2013;12:191–204.

    Article  CAS  PubMed  Google Scholar 

  162. Fava M, Evins AE, Dorer DJ, Schoenfeld DA. The problem of the placebo response in clinical trials for psychiatric disorders: culprits, possible remedies, and a novel study design approach. Psychother Psychosom. 2003;72:115–27.

    Article  PubMed  Google Scholar 

  163. Johnson KM, Devine JM, Ho KF, Howard KA, Saretsky TL, Jamieson CA. Evidence to support Montgomery-Asberg Depression Rating Scale administration every 24 h to assess rapid onset of treatment response. J Clin Psychiatry. 2016;77:1681–86.

    Article  PubMed  Google Scholar 

  164. Amidfar M, Réus GZ, Quevedo J, Kim Y-K. The role of memantine in the treatment of major depressive disorder: clinical efficacy and mechanisms of action. Eur J Pharm. 2018;827:103–11.

    Article  CAS  Google Scholar 

  165. Zarate CA Jr, Mathews D, Ibrahim L, Chaves JF, Marquardt C, Ukoh I, et al. A randomized trial of a low-trapping nonselective N-methyl-D-aspartate channel blocker in major depression. Biol Psychiatry. 2013;74:257–64.

    Article  CAS  PubMed  Google Scholar 

  166. Sanacora G, Smith MA, Pathak S, Su HL, Boeijinga PH, McCarthy DJ, et al. Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects. Mol Psychiatry. 2014;19:978–85.

    Article  CAS  PubMed  Google Scholar 

  167. Sanacora G, Johnson MR, Khan A, Atkinson SD, Riesenberg RR, Schronen JP, et al. Adjunctive lanicemine (AZD6765) in patients with major depressive disorder and history of inadequate response to antidepressants: a randomized, placebo-controlled study. Neuropsychopharmacology. 2017;42:844–53.

    Article  CAS  PubMed  Google Scholar 

  168. Henter ID, Park LT, Zarate CA Jr. Novel glutamatergic modulators for the treatment of mood disorders: current status. CNS Drugs. 2021;35:527–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Nutt JG, Gunzler SA, Kirchhoff T, Hogarth P, Weaver JL, Krams M, et al. Effects of a NR2B selective NMDA glutamate antagonist, CP‐101,606, on dyskinesia and parkinsonism. Mov Disord. 2008;23:1860–66.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Kadriu B, Musazzi L, Henter ID, Graves M, Popoli M, Zarate CA Jr. Glutamatergic neurotransmission: pathway to developing novel rapid-acting antidepressant treatments. Int J Neuropsychopharmacol. 2019;22:119–35.

    Article  CAS  Google Scholar 

  171. Kuppili PP, Venon V, Sathyanarayanan G, Sarkar S, Andrade C. Efficacy of adjunctive D-Cycloserine for the treatment of schizophrenia: a systematic review and meta-analysis of randomized controlled trials. J Neural Transm. 2021;128:253–62.

    Article  CAS  PubMed  Google Scholar 

  172. Cole J, Sohn MN, Harris AD, Bray SL, Patten SB, McGirr A. Efficacy of adjunctive D-cycloserine to intermittent theta-burst stimulation for major depressive disorder: a randomized clinical trial. JAMA Psychiatry. 2022;79:1153–61.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Farrell LJ, Waters AM, Tiralongo E, Mathieu S, McKenzie M, Garbharran V, et al. Efficacy of D-cycloserine augmented brief intensive cognitive-behavioural therapy for paediatric obsessive-compulsive disorder: a randomised clinical trial. Depress Anxiety. 2022;39:461–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Inslicht SS, Niles AN, Metzler TJ, Lipshitz SAL, Otte C, Milad MR, et al. Randomized controlled experimental study of hydrocortisone and D-cycloserine effects on fear extinction in PTSD. Neuropsychopharmacology. 2022;47:1945–52.

    Article  CAS  PubMed  Google Scholar 

  175. Chen M-H, Cheng C-M, Gueorguieva R, Lin W-C, Li C-T, Hong C-J, et al. Maintenance of antidepressant and antisuicidal effects by D-cycloserine among patients with treatment-resistant depression who responded to low-dose ketamine infusion: a double-blind randomized placebo–control study. Neuropsychopharmacology. 2019;44:2112–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Nagele P, Zorumski CF, Conway C. Exploring nitrous oxide as treatment for mood disorders: basic concepts. J Clin Psychopharmacol. 2018;38:144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Nagele P, Duma A, Kopec M, Gebara MA, Parsoei A, Walker M, et al. Nitrous oxide for treatment-resistant major depression: a proof-of-concept trial. Biol Psychiatry. 2015;78:10–18.

    Article  CAS  PubMed  Google Scholar 

  178. Varias A, van Roessel P, Parsiani M, Filippou-Frye M, Neylan TC, Nagele P, et al. Does nitrous oxide help veterans with post-traumatic stress disorder (PTSD)? A case series. J Clin Psychiatry. 2020;81:20I13393.

    Article  Google Scholar 

  179. Kim WSH, Dimick MK, Omrin D, Mitchell RHB, Riegart D, Levitt A, et al. Proof‐of‐concept randomized controlled trial of single‐session nitrous oxide for refractory bipolar depression: focus on cerebrovascular target engagement. Bipolar Disord. 2022;25:221–32.

  180. Nagele P, Palanca BJ, Gott B, Brown F, Barnes L, Nguyen T, et al. A phase 2 trial of inhaled nitrous oxide for treatment-resistant major depression. Sci Transl Med. 2021;13:eabe1376.

    Article  CAS  PubMed  Google Scholar 

  181. Ghaemi N, Sverdlov A, Shelton R, Litman R. Efficacy and safety of mij821 in patients with treatment-resistant depression: results from a randomized, placebo-controlled, proof-of-concept study. Eur Psychiatry. 2021;64:S334–35.

    Article  PubMed Central  Google Scholar 

  182. Bettini E, Stahl SM, De Martin S, Mattarei A, Sgrignani J, Carignani C, et al. Pharmacological comparative characterization of REL-1017 (esmethadone-HCl) and other NMDAR channel blockers in human heterodimeric N-methyl-D-aspartate receptors. Pharmaceuticals. 2022;15:997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. De Martin S, Gabbia D, Folli F, Bifari F, Fiorina P, Ferri N, et al. REL-1017 (esmethadone) increases circulating BDNF levels in healthy subjects of a Phase 1 clinical study. Front Pharmacol. 2021;12:671859.

  184. Fava M, Stahl S, Pani L, De Martin S, Pappagallo M, Guidetti C, et al. REL-1017 (Esmethadone) as adjunctive treatment in patients with major depressive disorder: a phase 2a randomized double-blind trial. Am J Psychiatry. 2022;179:122–31.

    Article  Google Scholar 

  185. Guidetti C, Fava M, Pani L, Pappagallo M, Serra G, DeMartin S, et al. A Phase 2a double-blind randomized trial of REL-1017 (Esmethadone) in patients with MDD: Analysis of subscales from the symptoms of depression questionnaire. CNS Spectr. 2022;27:235.

    Article  Google Scholar 

  186. Iosifescu DV, Jones M, O’Gorman C, Streicher C, Feliz S, Fava M, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: A phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83:21m14345.

    Article  PubMed  Google Scholar 

  187. Stahl SM. Dextromethorphan/bupropion: a novel oral NMDA (N-methyl-d-aspartate) receptor antagonist with multimodal activity. CNS Spectr. 2019;24:461–66.

    Article  PubMed  Google Scholar 

  188. Lepkowsky CM. Auvelity as a potential treatment for Alzheimer’s disease. Med Res Arch. 2022;10. https://doi.org/10.18103/mra.v10i10.3206.

  189. Machado-Vieira R, Manji HK, Zarate CA. The role of the tripartite glutamatergic synapse in the pathophysiology and therapeutics of mood disorders. Neuroscientist. 2009;15:525–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011;475:91–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharm Toxicol. 2010;50:295–322.

    Article  CAS  Google Scholar 

  192. Zhang K, Xu T, Yuan Z, Wei Z, Yamaki VN, Huang M, et al. Essential roles of AMPA receptor GluA1 phosphorylation and presynaptic HCN channels in fast-acting antidepressant responses of ketamine. Sci Signal. 2016;9:ra123.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Zanos P, Highland JN, Stewart BW, Georgiou P, Jenne CE, Lovett J, et al. (2R,6R)-hydroxynorketamine exerts mGlu2 receptor-dependent antidepressant actions. Proc Natl Acad Sci USA. 2019;116:6441–50.

  194. Watanabe M, Marcy B, Hiroki A, Watase H, Kinoshita K, Iijima M, et al. Evaluation of the safety, tolerability, and pharmacokinetic profiles of TP0473292 (TS-161), a prodrug of a novel orthosteric mGlu2/3 receptor antagonist TP0178894, in healthy subjects and its antidepressant-like effects in rodents. Neuropsychopharmacology. 2022;25:106–17.

    Article  CAS  Google Scholar 

  195. Umbricht D, Niggli M, Sanwald-Ducray P, Deptula D, Moore R, Grünbauer W, et al. Randomized, double-blind, placebo-controlled trial of the mGlu2/3 negative allosteric modulator decoglurant in partially refractory major depressive disorder. J Clin Psychiatry. 2020;81:18m12470.

    Article  PubMed  Google Scholar 

  196. Agnieszka P-P, Karolina P, Andrzej P. Role of AMPA receptor stimulation and TrkB signaling in the antidepressant-like effect of ketamine co-administered with a group II mGlu receptor antagonist, LY341495, in the forced swim test in rats. Behav Pharm. 2019;30:471–7.

    Article  CAS  Google Scholar 

  197. Rafało-Ulińska A, Brański P, Pałucha-Poniewiera A. Combined administration of (R)-ketamine and the mGlu2/3 receptor antagonist LY341495 induces rapid and sustained effects in the CUMS model of depression via a TrkB/BDNF-dependent mechanism. Pharmaceuticals. 2022;15:125.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Kadriu B, Musazzi L, Johnston J, Kalynchuk LE, Caruncho HJ, Popoli M, et al. Positive AMPA receptor modulation in the treatment of neuropsychiatric disorders: a long and winding road. Drug Discov Today. 2021;26:2816–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Ornstein PL, Zimmerman DM, Arnold MB, Bleisch TJ, Cantrell B, Simon R, et al. Biarylpropylsulfonamides as novel, potent potentiators of 2-amino-3-(5-methyl-3-hydroxyisoxazol-4-yl)-propanoic acid (AMPA) receptors. J Med Chem. 2000;43:4354–58.

    Article  CAS  PubMed  Google Scholar 

  200. O’Neill MJ, Witkin JM. AMPA receptor potentiators: application for depression and Parkinson’s disease. Curr Drug Targets. 2007;8:603–20.

    Article  PubMed  Google Scholar 

  201. Chappell AS, Gonzales C, Williams J, Witte MM, Mohs RC, Sperling R. AMPA potentiator treatment of cognitive deficits in Alzheimer disease. Neurology. 2007;68:1008–12.

    Article  CAS  PubMed  Google Scholar 

  202. Trzepacz PT, Cummings J, Konechnik T, Forrester TD, Chang C, Dennehy EB, et al. Mibampator (LY451395) randomized clinical trial for agitation/aggression in Alzheimer’s disease. Int Psychogeriatr. 2013;25:707–19.

    Article  PubMed  Google Scholar 

  203. Kays JL, Hurley RA, Taber KH. The dynamic brain: neuroplasticity and mental health. J Neuropsychiatry Clin Neurosci. 2012;24:118–24.

    Article  PubMed  Google Scholar 

  204. Gandal MJ, Sisti J, Klook K, Ortinski PI, Leitman V, Liang Y, et al. GABAB-mediated rescue of altered excitatory-inhibitory balance, gamma synchrony and behavioral deficits following constitutive NMDAR-hypofunction. Transl Psychiatry. 2012;2:e142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Nugent AC, Ballard ED, Gould TD, Park LT, Moaddel R, Brutsche NE, et al. Ketamine has distinct electrophysiological and behavioral effects in depressed and healthy subjects. Mol Psychiatry. 2019;24:1040–52.

    Article  CAS  PubMed  Google Scholar 

  206. Shaw AD, Saxena N, Jackson LE, Hall JE, Singh KD, Muthukumaraswamy SD. Ketamine amplifies induced gamma frequency oscillations in the human cerebral cortex. Eur Neuropsychopharmacol. 2015;25:1136–46.

    Article  CAS  PubMed  Google Scholar 

  207. Tiger M, Veldman ER, Ekman C-J, Halldin C, Svenningsson P, Lundberg J. A randomized placebo-controlled PET study of ketamine´ s effect on serotonin1B receptor binding in patients with SSRI-resistant depression. Transl Psychiatry. 2020;10:159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Holmes SE, Finnema SJ, Naganawa M, DellaGioia N, Holden D, Fowles K, et al. Imaging the effect of ketamine on synaptic density (SV2A) in the living brain. Mol Psychiatry. 2022;27:2273–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Abdallah CG, Averill LA, Collins KA, Geha P, Schwartz J, Averill CL, et al. Ketamine treatment and global brain connectivity in major depression. Neuropsychopharmacology. 2017;42:1210–19.

    Article  CAS  PubMed  Google Scholar 

  210. Kraus C, Mkrtchian A, Kadriu B, Nugent AC, Zarate CA Jr, Evans JW. Evaluating global brain connectivity as an imaging marker for depression: influence of preprocessing strategies and placebo-controlled ketamine treatment. Neuropsychopharmacology. 2020;45:982–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Siegel JS, Palanca BJA, Ances BM, Kharasch ED, Schweiger JA, Yingling MD, et al. Prolonged ketamine infusion modulates limbic connectivity and induces sustained remission of treatment-resistant depression. Psychopharmacology. 2021;238:1157–69.

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Reed JL, Nugent AC, Furey ML, Szczepanik JE, Evans JW, Zarate CA Jr. Effects of ketamine on brain activity during emotional processing: diferential findings in depressed versus healthy control participants. Biol Psychiatry Cogn Neurosci Neuroimaging. 2019;4:610–18.

    PubMed  PubMed Central  Google Scholar 

  213. McMillan R, Forsyth A, Campbell D, Malpas G, Maxwell E, Dukart J, et al. Temporal dynamics of the pharmacological MRI response to subanaesthetic ketamine in healthy volunteers: a simultaneous EEG/fMRI study. J Psychopharmacol. 2019;33:219–29.

    Article  CAS  PubMed  Google Scholar 

  214. Zavaliangos-Petropulu A, Al-Sharif NB, Taraku B, Leaver AM, Sahib AK, Espinoza RT, et al. Neuroimaging-derived biomarkers of the antidepressant effects of ketamine. Biol Psychiatry Cogn Neurosci Neuroimaging. 2023;8:361–86.

  215. Kadriu B, Ballard ED, Henter ID, Murata S, Gerlus N, Zarate CAJ. Neurobiological biomarkers of response to ketamine. Adv Pharm. 2020;89:195–235.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank the 7SE research unit and staff for their support.

Funding

Funding for this work was provided by the Intramural Research Program at the National Institute of Mental Health, National Institutes of Health (IRP-NIMH-NIH; ZIAMH002857). Additional funding was provided by a NARSAD Young Investigator Award to BK. The NIMH and NARSAD had no further role in study design; in the collection, analysis, or interpretation of data; in the writing of the report; or in the decision to submit the paper for publication. The work was completed as part of the authors’ official duties as Government employees. The views expressed do not necessarily reflect the views of the NIH, the Department of Health and Human Services, or the United States Government.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed equally to the conceptualization of the manuscript, literature search, generation of the figure and table, writing, and revision of this manuscript. All authors approved the final version of the paper.

Corresponding author

Correspondence to Jenessa N. Johnston.

Ethics declarations

Competing interests

CAZ is listed as a co-inventor on a patent for the use of ketamine in major depression and suicidal ideation; as a co-inventor on a patent for the use of (2R,6R)-hydroxynorketamine, (S)-dehydronorketamine, and other stereoisomeric dehydroxylated and hydroxylated metabolites of (R,S)-ketamine metabolites in the treatment of depression and neuropathic pain; and as a co-inventor on a patent application for the use of (2R,6R)-hydroxynorketamine and (2S,6S)-hydroxynorketamine in the treatment of depression, anxiety, anhedonia, suicidal ideation, and post-traumatic stress disorders. He has assigned his patent rights to the U.S. government but will share a percentage of any royalties that may be received by the government. BK is now a full-time employee and shareholder of Jazz Pharmaceuticals. All other authors have no conflict of interest to disclose, financial or otherwise.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Johnston, J.N., Kadriu, B., Kraus, C. et al. Ketamine in neuropsychiatric disorders: an update. Neuropsychopharmacol. 49, 23–40 (2024). https://doi.org/10.1038/s41386-023-01632-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41386-023-01632-1

  • Springer Nature Switzerland AG

This article is cited by

Navigation