Open access peer-reviewed chapter

Emergence of Ketamine as a Rapid Acting Antidepressant: Mechanistic Insights and Future Directions

Written By

Atamjit Singh and Preet Mohinder Singh Bedi

Submitted: 12 July 2021 Reviewed: 02 August 2021 Published: 10 September 2021

DOI: 10.5772/intechopen.99765

From the Edited Volume

Ketamine Revisited - New Insights into NMDA Inhibitors

Edited by Nieves Saiz-Sapena and Manuel Granell-Gil

Chapter metrics overview

300 Chapter Downloads

View Full Metrics

Abstract

Ketamine is a phencyclidine derivative and N-methyl-D-aspartate receptor antagonist, widely popular as a dissociative anesthetic. Its use as an anesthetic in humans was progressively fallen out due to its associated adverse effects and the emergence of newer and safer anesthetics. In recent few decades, various reports related to its efficacy in the treatment of resistant depression with anti-suicidal potential draw significant attention from researchers around the globe. The rapid clinical effect of ketamine within hours as compared to traditional antidepressants that take several weeks makes it a hot topic in antidepressant research. Studies conducted in the recent past suggest its mechanism of action through glutamate modulation via receptors like NMDA, AMPA as well as downregulation of BDNF etc. This chapter will shed light on the various mechanisms of ketamine related to antidepressant activity. Along with that its pharmacokinetics, toxicology and ongoing clinical trials will also be discussed.

Keywords

  • ketamine
  • depression
  • antidepressant
  • NMDA
  • BDNF

1. Introduction

From last few decades with rapid development and modernization, significant improvements in the lifestyle of humans has been observed but with pros there are associated cons and so is major depressive disorder (MDD) which is affecting teenagers to adults and majorly observed in young working professionals. It is emerging as major contributor in global disease burden and reported as the second leading causes for disability [1]. According to the study conducted by mental health in Canada, MDD has lifetime prevalence of 11.3% [2]. Besides being a major challenge for healthcare system its pathophysiology is still not uncovered completely. One hypothesis based on monoamines suggest that it may resulted from functional deficiency of neurotransmitters named serotonin and/or noradrenaline which is widely utilized for categorization of antidepressant drugs [3]. But conflict is also standstill with the time frame of the effect and dose administration as clinical symptoms are observed after several weeks from the onset of therapy and only half are noted to have actual clinical response [4, 5, 6, 7]. Apart from that one-third patients suffers from treatment resistant depression (TRD) that are nonresponsive to currently approved medications [8]. Non-responsiveness of currently available therapy especially for TRD arise the emergency need of more effective and safer antidepressant therapy.

Ketamine is a phencyclidine derivative and N-methyl-D-aspartate (NMDA) receptor antagonist, widely popular as a dissociative anesthetic. Ketamine was first reported for its efficacy in depression in year 2000, when sub-anesthetic intravenous dose of ketamine rapidly reduced the symptoms of MDD and effect continued up to 72 hours [9]. Taking lead from this, further clinical trials were conducted which showcase its efficacy in TRD patients with 60–70% response rate [10, 11, 12, 13, 14]. Onset of action was reported within 2–4 hours and last for 1 week with singe infusion while repeated infusions have effect up to 18–19 days. Clinical data also suggest the responsiveness of ketamine up to 44% on patients with comorbidities and ultra-resistant depression [15, 16]. In addition to this ketamine has been reported for its anti-suicidal and anti-anhedonic properties [14, 17, 18]. All this reports points toward the different mechanism of ketamine form traditional antidepressants.

Advertisement

2. Basic chemistry, pharmacology and pharmacokinetics of ketamine

Recently discovered antidepressant and anti-suicidal action of ketamine significantly attracted the researchers working in the field of psychiatry [9, 11, 19]. Ketamine is a phencyclidine derivative and a mixture of R(−) and S(+) enantiomers. Both R(−) and S(+) enantiomers has been explored widely and it was observed that S(+) enantiomer has higher potency than R(−) enantiomer (R-ketamine) for phencyclidine site on glutamate NMDA receptor along with stronger analgesic activity [20, 21, 22, 23, 24]. Inspired form these outputs, S(+) enantiomer also known as esketamine is now under investigations for antidepressant potential [25]. However conflict between these two is also exist with the side effects profile of both enantiomers related to dissociation, psychoses and cognition [26]. Reports suggest the rapid onset of antidepressant effects with R-ketamine but higher side effects than esketamine [27, 28, 29, 30, 31, 32, 33, 34]. Ketamine undergo metabolism through CYP2B6- and CYP3A4-mediated N-demethylation resulting norketamine which further catabolized into hydroxynorketamines (HNKs) and dehyronorketamine (Figure 1). Investigations was also carried out on metabolites of ketamine. 2R,6R-HNK has been observed to have antidepressant like efficacy with nil side effects on rat models while several contradictory reports are also available [35, 36, 37, 38, 39, 40, 41, 42, 43]. Specifically, metabolite of esketamine i.e. S-norketamine showed antidepressant like properties with lesser side effects as with esketamine [44]. When talk about bioavailability, ketamine has varying bioavailability profile with different routes i.e. 100% with intravenous, 45% with intranasal, 30% with sublingual, 20% with oral, 93% with intramuscular while 30% with rectal route [24, 30, 44].

Figure 1.

General layout of metabolic pathway of ketamine showcasing stereoseletive metabolism through various cytochrome P450 enzymes.

Advertisement

3. Overview of the status of clinical trials with ketamine and its enantiomers

Report on antidepressant efficacy of ketamine by Berman group in 2000 [9] initiated series of studies related to antidepressant activity of ketamine all around the globe. Multiple meta-analysis now established the candidature of ketamine against major depressive episodes in both bipolar as well as unipolar depression while efficacy was higher in unipolar as compared to bipolar depression [45, 46, 47, 48, 49, 50]. In addition to this, numerous studies reported its effect last up to a week only for unipolar while it is up to 3–4 days in case of bipolar depression [46, 47, 49]. Randomized Controlled trials (RCT) exist in which effect of repeated infusions of ketamine for depression is studied but there is still lack of long term trial [51, 52, 53]. Studies on different routes of administration were also conducted that majorly include intranasal, sublingual and intramuscular [54, 55, 56, 57]. In fact intranasal esketaminerecently got FDA clearance for TRD which was based on three acute-phase and two maintenance phase studies. These acute studies were conducted on severely depressed patients [58]. Maintenance trials were conducted up to 88 weeks where patient was administered esketamine weekly or every second week showcase reduced after relapse risk and also assured safety up to a year [59, 60]. A phase three trial consisted of 200 patients suggest the significant improvements in depression with ketamine adjuvant to an antidepressant [61]. There is another 5 year ongoing trial by Janssen for safety [62]. Keeping in view the antidepressant efficacy if R-ketamine, a phase I trial was started by Perception Pharmaceuticals but results are not processed yet [28].

Advertisement

4. Mechanistic insight into the antidepressant activity of ketamine

4.1 AMPA, BDNF and mTOR

Glutamate is one of the major excitatory neurotransmitters in central nervous system of human body that mainly acts on NMDA, ionotropic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (co-localized with NMDA) and metabotropic glutamate receptors. Glutamate activates AMPA receptors at synaptic cleft, which permit the entry of sodium ions into postsynaptic membrane. Entry of sodium ions results in depolarization of postsynaptic membrane that cause removal of NMDA receptor channel voltage-dependent magnesium ion block that activate NMDA receptor which allow the entry of sodium as well as calcium ions. Ketamine is a well-established non-competitive type NMDA receptor antagonist. Brain-derived neurotrophic factor (BDNF) and mTOR are two major proteins that are suspected to be involved in mechanistic window of ketamine. BDNF is a growth factor protein in central nervous system that promote neurogenesis and synaptogenesis along with support in survival of existing neurons. On the other hand, mTOR is suggested to have major role in neuronal development and circuit formation. mTOR further made two sub complexes known as mTOR complex 1 (mTORC1) and mTOR, from which mTORC1 is a target of ketamine [63, 64].

It has been observe that glutamatergic neurotransmission is deregulated in MDD and enhanced levels of glutamate levels in serum and plasma were observed in patient’s dealing with MDD that why plasma glutamate levels are directly correlated with severity of depression [65, 66, 67, 68]. Enhanced glutamate cause by loss of glial cells in MDD increases extra synaptic glutamate levels that suppressglutamatergic neurotransmission via activation of metabotropic glutamate receptor 2 (mGluR2) autoreceptors. A study suggest that change in depression symptoms by non-ketamine NMDA receptor antagonists like traxoprodil, lanicemine and rapastinel was much lower ass compared to ketamine [34, 69, 70, 71]. Ketamine good antagonistic activity for NMDA receptors present on γ-aminobutyric acid (GABA) that prevent activation of GABA interneurons resulting in downstream disinhibition of glutamatergic neurons that cause glutamate surge. Elevated levels of glutamate initiates activation of postsynaptic AMPA receptors that potentiate BDNF andmTORC1 signaling pathways. Ketamine demonstrated activate glutamate release and transmission in rat prefrontal cortex (RPC) [72]. Ketamine was also observed to enhance AMPA-evoked electrophysiological responses in the rat hippocampus and medial PFC pointing toward the involvement of ketamine in AMPA receptor transmission [73, 74, 75, 76, 77]. In a mouse model, ketamine was observed to increase the expression levels of two subunits of AMPA receptor known as GluA1 and GluA2 [34, 78].

Increased levels of BDNF and mTOR in rat hippocampus were observed within 30 minutes of treatment with ketamine [73, 79, 80]. Important to mention here that analgesic tramadol enhanced the effect of ketamine on force swim test along with upregulation of mTOR in the PFC and hippocampus of rat [81]. It is interesting to observe that increased BDNF and mTOR levels in hippocampal and RFC are controlled by AMPA because in a study treatment with AMPA receptor antagonist increased forced-swim test immobility time with reduced levels of BDNF and mTOR while with agonist immobility time reduced along with increased levels of both BDNF and mTOR [82]. Reports were also observed that suggest the nullification of antidepressant activity of ketamine with pre-treatment of rapamycin an mTORC1 inhibitor [83].

Numerous reports are present in the literature suggesting the possibility of ketamine’s antidepressant activity via BDNF. No antidepressant activity was observed on treatment of ketamine in genetically modified mice lacking BDNF [73]. It is proposed that antagonism of NMDA through ketamine deactivates the eukaryotic elongation of factor 2 (eEF2) kinase that de-supress the translation of BDNF. Mice having Val66Met single-nucleotide polymorphism in BDNF gene showed impairment in BDNF release and mRNA trafficking. Administration of ketamine in these mice showed reduced antidepressant activity [84]. Reversal of anhedonicbehaviour with ketamine was observed in rats with chronic mild stress along with complete restoration of dendritic atrophy and dendritiv BDNF mRNA trafficking [85]. In social defeat stress model of mice, ketamine lessen reduction in BDNF, spine density of dendrites, synaptogenesis markers (GluA1 and PSD-95) in PFC, CA3 and dentate gyrus region of hippocampus at 8th day of treatment [86]. Elevated levels of BDNF were supposed to be associated with the lower severity of depression like symptoms on rating scale [87, 88]. A study carried out on three depressed patients, suggest their response to ketamine and have increased levels of plasma mTOR expression and eEF2 phosphorylaton [89]. It is worth to note that in a trial conducted on 20 patients, pre-treatment with rapamycin tripled the response rate after 2 weeks from treatment thus may be due to targeting of rapamycin on neuroinflammation through its immunisupressant activity or may be due to promotion of haemostatsis of synaptic density (Figure 2) [90].

Figure 2.

Flow diagram of antidepressant activity of ketamine. (1) ketamine binds with N-methyl-d-aspartate receptors (NMDARs) and reduce excitability of γ-aminobutyric acid (GABA) ergic interneurons that results, (2) non-inhibition of glutamatergic neurons, (3) that further increase glutamate release which binds with α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors resulting inflow of sodium and calcium into cell, (4) cause activation of voltage gated calcium channels, (5) that further triggers the release of brain-derived neurotrophic factor (BDNF) into glutamate synapse. (6) BDNF from synapse binds with tropomyosin receptor kinase B (TrkB) resulting activation of MEK–ERK and PI3K-Akt signaling cascades that converge on to mTOR lead to (7) increased synaptic protein translation. (8) increased proteins in synapse lead to increased AMPAR-mediated synaptic transmission causing elevated synaptogenesis. All these events are hypothesized to restore disrupted connectivity between key brain regions and can be the possible reason of rapid and sustained antidepressant action of ketamine.

4.2 D-serine

D-serine is a potential co-agonist at NMDA receptor which is a possible biomarker in depression. Numerous studies highlighted the abnormality of D-serine levels in depression highlighting the antidepressant properties of D-serine [91, 92, 93, 94, 95]. Ketamine was found to inhibitor the transport of D-serine while ketamine metabolites were observed to decrease intracellular (PC-12 cells) concentrations of D-serine thus increasing plasma D-serine levels which is possible prediction related to its to antidepressant action [96, 97, 98, 99].

4.3 Opioid system

Ketamine also have capability to bind with opioid receptors (mu, delta and kappa), monoaminergic receptors and transporters, and muscarinic and nicotinic cholinergic receptors [100]. Proposition is made that anti-suicidal as well as antidepressant actions of ketamine is related to the opioid system which is confirmed from the pre-treatment of naltrexone after that antidepressant effect was attenuated in patients [100, 101]. However many discrepancies are also exist along with [102, 103] because buprenorphine and methadone both are agonists to the opioid receptors and does not have any effect on antidepressant properties of ketamine [103]. These results rebels the role of opioid system in ketamine’s antidepressant effects. Thus role of opioid in ketamine’s antidepressant effects is yet unclear and controversial.

Advertisement

5. Future trends

With unique mechanism of action as compared to traditional antidepressants along with anti-suicidal properties, ketamine successfully attracted the researchers and physiologists toward itself in last two decades. However large mechanism of actions are still need to uncover thus it will be continue to be a hot topic and active area of research in psychiatry. There if a dire need to investigate the appropriate safety to efficacy ration of ketamine in depression therapy along with establishment of appropriate regimens for maintenance of therapy and discontinuation too. Reliable biomarkers are also needed to properly predict the response and adverse effects of ketamine. Numerous reports are also present in literature that caution the utilization of ketamine as an antidepressant in clinical practice [76, 104, 105, 106, 107, 108]. Keeping these thing apart, currently ketamine is emerging as a promising approach for treatment of patients suffering from TRD. Ketamine and its related neurochemical biomarkers can act as leads for development of future antidepressants.

Advertisement

6. Conclusion

Rapid antidepressant effect of ketamine depression therapy and important discovery in depression research. Its efficacy against TRD and anti-suicidal potential is a boon in depression research but at the same time its negative side effects and potential for being abuse is not to be neglected. However pathways like BDNF, mTOR, AMPA along D-serine and opioid receptors provided sufficient understanding but large portion of its mechanisms are still need to uncover. Even some studies create conflict to each other which is needed to be resolved. Overall analysis suggest that there is an important need to discover all aspects of ketamine in depression therapy to efficient use of this drug as an antidepressant in clinical practice. Moreover, ketamine can act as a lead for the development of new class of rapidly acting future antidepressant agents.

Advertisement

Acknowledgments

The authors are also thankful to Guru Nanak Dev University, Amritsar for providing various facilities to carry out the work.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Ferrari AJ, Charlson FJ, Norman RE, Patten SB, Freedman G, Murray CJ, Vos T, Whiteford HA. Burden of depressive disorders by country, sex, age, and year: Findings from the global burden of disease study 2010. PLoS medicine. 2013 Nov 5;10(11):e1001547.
  2. 2. Patten SB, Williams JV, Lavorato DH, Wang JL, McDonald K, Bulloch AG. Descriptive epidemiology of major depressive disorder in Canada in 2012. The Canadian Journal of Psychiatry. 2015 Jan;60(1):23-30.
  3. 3. Matveychuk D, Thomas RK, Swainson J, Khullar A, MacKay MA, Baker GB, Dursun SM. Ketamine as an antidepressant: Overview of its mechanisms of action and potential predictive biomarkers. Therapeutic Advances in Psychopharmacology. 2020 May;10:2045125320916657.
  4. 4. Hirschfeld RM. History and evolution of the monoamine hypothesis of depression. Journal of clinical psychiatry. 2000 Mar 31;61(6):4-6.
  5. 5. Liu B, Liu J, Wang M, Zhang Y, Li L. From serotonin to neuroplasticity: Evolvement of theories for major depressive disorder. Frontiers in cellular neuroscience. 2017 Sep 28;11:305.
  6. 6. Undurraga J, Baldessarini RJ. Randomized, placebo-controlled trials of antidepressants for acute major depression: Thirty-year meta-analytic review. Neuropsychopharmacology. 2012 Mar;37(4):851-864.
  7. 7. Fava M, Davidson KG. Definition and epidemiology of treatment-resistant depression. Psychiatric Clinics of North America. 1996 Jun 1;19(2):179-200.
  8. 8. Nemeroff CB. Prevalence and management of treatment-resistant depression. Journal of Clinical Psychiatry. 2007 Jul 16;68(8):17.
  9. 9. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH. Antidepressant effects of ketamine in depressed patients. Biological psychiatry. 2000 Feb 15;47(4):351-354.
  10. 10. Murrough JW, Iosifescu DV, Chang LC, Al Jurdi RK, Green CE, Perez AM, Iqbal S, Pillemer S, Foulkes A, Shah A, Charney DS. Antidepressant efficacy of ketamine in treatment-resistant major depression: A two-site randomized controlled trial. American Journal of Psychiatry. 2013 Oct;170(10):1134-1142.
  11. 11. Zarate CA, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, Charney DS, Manji HK. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Archives of general psychiatry. 2006 Aug 1;63(8):856-864.
  12. 12. Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, Kammerer WA, Quezado Z, Luckenbaugh DA, Salvadore G, Machado-Vieira R. a randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Archives of general psychiatry. 2010 Aug 1;67(8):793-802.
  13. 13. Coyle CM, Laws KR. The use of ketamine as an antidepressant: A systematic review and meta-analysis. Human Psychopharmacology: Clinical and Experimental. 2015 May;30(3):152-163.
  14. 14. Wilkinson ST, Ballard ED, Bloch MH, Mathew SJ, Murrough JW, Feder A, Sos P, Wang G, Zarate Jr CA, Sanacora G. The effect of a single dose of intravenous ketamine on suicidal ideation: A systematic review and individual participant data meta-analysis. American journal of psychiatry. 2018 Feb 1;175(2):150-158.
  15. 15. Thomas RK, Baker G, Lind J, Dursun S. Rapid effectiveness of intravenous ketamine for ultraresistant depression in a clinical setting and evidence for baseline anhedonia and bipolarity as clinical predictors of effectiveness. Journal of psychopharmacology. 2018 Oct;32(10):1110-1117.
  16. 16. Thomas, R., Baker, G. and Dursun, S., 2017, November. Rapid Efficacy and Antisuicidal Actions of Intravenous Ketamine for Ultraresistant Depression in a Clinical Setting: A Retrospective, Database Study. In Neuropsychopharmacology (Vol. 43, Pp. S174-S174). Macmillan Building, 4 Crinan St, London N1 9xw, England: Nature Publishing Group.
  17. 17. Grunebaum MF, Galfalvy HC, Choo TH, Keilp JG, Moitra VK, Parris MS, Marver JE, Burke AK, Milak MS, Sublette ME, Oquendo MA. Ketamine for rapid reduction of suicidal thoughts in major depression: A midazolam-controlled randomized clinical trial. American Journal of Psychiatry. 2018 Apr 1;175(4):327-335.
  18. 18. Zanos P, Thompson SM, Duman RS, Zarate CA, Gould TD. Convergent mechanisms underlying rapid antidepressant action. CNS drugs. 2018 Mar;32(3):197-227.
  19. 19. DiazGranados N, Ibrahim LA, Brutsche NE, Ameli R, Henter ID, Luckenbaugh DA, Machado-Vieira R, Zarate Jr CA. Rapid resolution of suicidal ideation after a single infusion of an N-methyl-D-aspartate antagonist in patients with treatment-resistant major depressive disorder. The Journal of clinical psychiatry. 2010 Jul 13;71(12):1605-1611.
  20. 20. Muller J, Pentyala S, Dilger J, Pentyala S. Ketamine enantiomers in the rapid and sustained antidepressant effects. Therapeutic advances in psychopharmacology. 2016 Jun;6(3):185-192.
  21. 21. Pfenninger EG, Durieux ME, Himmelseher S. Cognitive impairment after small-dose ketamine isomers in comparison to equianalgesic racemic ketamine in human volunteers. The Journal of the American Society of Anesthesiologists. 2002 Feb 1;96(2):357-366.
  22. 22. Andrade C. Ketamine for depression, 3: Does chirality matter?. The Journal of clinical psychiatry. 2017 Jun 28;78(6):e674-e676.
  23. 23. Muller J, Pentyala S, Dilger J, Pentyala S. Ketamine enantiomers in the rapid and sustained antidepressant effects. Therapeutic advances in psychopharmacology. 2016 Jun;6(3):185-192.
  24. 24. Peltoniemi MA, Hagelberg NM, Olkkola KT, Saari TI. Ketamine: A review of clinical pharmacokinetics and pharmacodynamics in anesthesia and pain therapy. Clinical pharmacokinetics. 2016 Sep;55(9):1059-1077.
  25. 25. Kim J, Farchione T, Potter A, Chen Q, Temple R. Esketamine for treatment-resistant depression-first FDA-approved antidepressant in a new class. N Engl J Med. 2019 Jul 4;381(1):1-4.
  26. 26. Hashimoto K. Rapid-acting antidepressant ketamine, its metabolites and other candidates: A historical overview and future perspective. Psychiatry and clinical neurosciences. 2019 Oct;73(10):613-627.
  27. 27. Lane RM, Baker GB. Chirality and drugs used in psychiatry: Nice to know or need to know?. Cellular and molecular neurobiology. 1999 Jun;19(3):355-372.
  28. 28. Hashimoto K, Yang C. Is (S)-norketamine an alternative antidepressant for esketamine?. European archives of psychiatry and clinical neuroscience. 2019 Oct;269(7):867-868.
  29. 29. Zhang JC, Li SX, Hashimoto K. R (−)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacology Biochemistry and Behavior. 2014 Jan 1;116:137-141.
  30. 30. Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, Dong C, Hashimoto K. R-ketamine: A rapid-onset and sustained antidepressant without psychotomimetic side effects. Translational psychiatry. 2015 Sep;5(9):e632.
  31. 31. Yang C, Han M, Zhang JC, Ren Q, Hashimoto K. Loss of parvalbumin-immunoreactivity in mouse brain regions after repeated intermittent administration of esketamine, but not R-ketamine. Psychiatry research. 2016 May 30;239:281-283.
  32. 32. Fukumoto K, Toki H, Iijima M, Hashihayata T, Yamaguchi JI, Hashimoto K, Chaki S. Antidepressant potential of (R)-ketamine in rodent models: Comparison with (S)-ketamine. Journal of Pharmacology and Experimental Therapeutics. 2017 Apr 1;361(1):9-16.
  33. 33. Hashimoto K, Kakiuchi T, Ohba H, Nishiyama S, Tsukada H. Reduction of dopamine D 2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: A PET study in conscious monkeys. European archives of psychiatry and clinical neuroscience. 2017 Mar 1;267(2):173-176.
  34. 34. Yang C, Yang J, Luo A, Hashimoto K. Molecular and cellular mechanisms underlying the antidepressant effects of ketamine enantiomers and its metabolites. Translational psychiatry. 2019 Nov 7;9(1):1-1.
  35. 35. Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, Alkondon M, Yuan P, Pribut HJ, Singh NS, Dossou KS. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016 May;533(7604):481-486.
  36. 36. Chaki S. Is metabolism of (R)-ketamine essential for the antidepressant effects?. International Journal of Neuropsychopharmacology. 2018 Feb;21(2):154-156.
  37. 37. Li L, Vlisides PE. Ketamine: 50 years of modulating the mind. Frontiers in human neuroscience. 2016 Nov 29;10:612.
  38. 38. Mion G, Villevieille T. Ketamine pharmacology: An update (pharmacodynamics and molecular aspects, recent findings). CNS neuroscience & therapeutics. 2013 Jun;19(6):370-380.
  39. 39. Zanos P, Highland JN, Liu X, Troppoli TA, Georgiou P, Lovett J, Morris PJ, Stewart BW, Thomas CJ, Thompson SM, Moaddel R. (R)-ketamine exerts antidepressant actions partly via conversion to (2R, 6R)-hydroxynorketamine, while causing adverse effects at sub-anaesthetic doses. British journal of pharmacology. 2019 Jul;176(14):2573-2592.
  40. 40. Fukumoto K, Fogaça MV, Liu RJ, Duman C, Kato T, Li XY, Duman RS. Activity-dependent brain-derived neurotrophic factor signaling is required for the antidepressant actions of (2R, 6R)-hydroxynorketamine. Proceedings of the National Academy of Sciences. 2019 Jan 2;116(1):297-302.
  41. 41. Chou D, Peng HY, Lin TB, Lai CY, Hsieh MC, Wen YC, Lee AS, Wang HH, Yang PS, Chen GD, Ho YC. (2R, 6R)-hydroxynorketamine rescues chronic stress-induced depression-like behavior through its actions in the midbrain periaqueductal gray. Neuropharmacology. 2018 Sep 1;139:1-2.
  42. 42. Xiong Z, Fujita Y, Zhang K, Pu Y, Chang L, Ma M, Chen J, Hashimoto K. Beneficial effects of (R)-ketamine, but not its metabolite (2R, 6R)-hydroxynorketamine, in the depression-like phenotype, inflammatory bone markers, and bone mineral density in a chronic social defeat stress model. Behavioural brain research. 2019 Aug 5;368:111904.
  43. 43. Yamaguchi JI, Toki H, Qu Y, Yang C, Koike H, Hashimoto K, Mizuno-Yasuhira A, Chaki S. (2 R, 6 R)-Hydroxynorketamine is not essential for the antidepressant actions of (R)-ketamine in mice. Neuropsychopharmacology. 2018 Aug;43(9):1900-1907.
  44. 44. Yang Y, Cui Y, Sang K, Dong Y, Ni Z, Ma S, Hu H. ketamine blocks bursting in the lateral habenula to rapidly relieve depression. Nature. 2018 Feb;554(7692):317-322.
  45. 45. Zhang K, Hashimoto K. An update on ketamine and its two enantiomers as rapid-acting antidepressants. Expert review of neurotherapeutics. 2019 Jan 2;19(1):83-92.
  46. 46. Kishimoto T, Chawla JM, Hagi K, Zarate CA, Kane JM, Bauer M, Correll CU. 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. Psychological medicine. 2016 May;46(7):1459-1472.
  47. 47. Lee EE, Della Selva MP, Liu A, Himelhoch S. Ketamine as a novel treatment for major depressive disorder and bipolar depression: A systematic review and quantitative meta-analysis. General hospital psychiatry. 2015 Mar 1;37(2):178-184.
  48. 48. McGirr A, Berlim MT, Bond DJ, Fleck MP, Yatham LN, Lam RW. A systematic review and meta-analysis of randomized, double-blind, placebo-controlled trials of ketamine in the rapid treatment of major depressive episodes. Psychological medicine. 2015 Mar;45(4):693-704.
  49. 49. Romeo B, Choucha W, Fossati P, Rotge JY. Meta-analysis of short-and mid-term efficacy of ketamine in unipolar and bipolar depression. Psychiatry research. 2015 Dec 15;230(2):682-688.
  50. 50. Fond G, Loundou A, Rabu C, Macgregor A, Lançon C, Brittner M, Micoulaud-Franchi JA, Richieri R, Courtet P, Abbar M, Roger M. Ketamine administration in depressive disorders: A systematic review and meta-analysis. Psychopharmacology. 2014 Sep;231(18):3663-3676.
  51. 51. Phillips JL, Norris S, Talbot J, Hatchard T, Ortiz A, Birmingham M, Owoeye O, Batten LA, Blier P. Single and repeated ketamine infusions for reduction of suicidal ideation in treatment-resistant depression. Neuropsychopharmacology. 2020 Mar;45(4):606-612.
  52. 52. Albott CS, Lim KO, Forbes MK, Erbes C, Tye SJ, Grabowski JG, Thuras P, Batres-y-Carr TM, Wels J, Shiroma PR. Efficacy, safety, and durability of repeated ketamine infusions for comorbid posttraumatic stress disorder and treatment-resistant depression. The Journal of clinical psychiatry. 2018 May 1;79(3):17m11634.
  53. 53. Zheng W, Zhou YL, Liu WJ, Wang CY, Zhan YN, Li HQ, Chen LJ, Li MD, Ning YP. Rapid and longer-term antidepressant effects of repeated-dose intravenous ketamine for patients with unipolar and bipolar depression. Journal of psychiatric research. 2018 Nov 1;106:61-68.
  54. 54. Lapidus KA, Levitch CF, Perez AM, Brallier JW, Parides MK, Soleimani L, Feder A, Iosifescu DV, Charney DS, Murrough JW. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biological psychiatry. 2014 Dec 15;76(12):970-976.
  55. 55. Gálvez V, Li A, Huggins C, Glue P, Martin D, Somogyi AA, Alonzo A, Rodgers A, Mitchell PB, Loo CK. Repeated intranasal ketamine for treatment-resistant depression—The way to go? Results from a pilot randomised controlled trial. Journal of Psychopharmacology. 2018 Apr;32(4):397-407.
  56. 56. Andrade C. Intranasal drug delivery in neuropsychiatry: Focus on intranasal ketamine for refractory depression. The Journal of clinical psychiatry. 2015 May 27;76(5):e628-e631.
  57. 57. Loo CK, Gálvez V, O'keefe E, Mitchell PB, Hadzi-Pavlovic D, Leyden J, Harper S, Somogyi AA, Lai R, Weickert CS, Glue P. Placebo-controlled pilot trial testing dose titration and intravenous, intramuscular and subcutaneous routes for ketamine in depression. ActaPsychiatricaScandinavica. 2016 Jul;134(1):48-56.
  58. 58. FDA. FDA report on esketamine for treatment resistant depression, https://www.fda.gov/media/121379/download (2021).
  59. 59. Daly EJ, Trivedi MH, Janik A, Li H, Zhang Y, Li X, Lane R, Lim P, Duca AR, Hough D, Thase ME. 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 Sep 1;76(9):893-903.
  60. 60. Wajs E, Aluisio L, Morrison R, Daly E, Lane R, Lim P, Holder R, Sanacora G, Young AH, Kasper S, Sulaiman AH. Long-Term Safety of Esketamine Nasal Spray plus Oral Antidepressant in Patients with Treatment-Resistant Depression: Phase 3, Open-Label, Safety and Efficacy Study (SUSTAIN-2). InPosterpresentado en the Annual Meeting of the American Society of Clinical Psychopharmacology, Miami, US 2018 May 29.
  61. 61. Popova V, Daly EJ, Trivedi M, Cooper K, Lane R, Lim P, Mazzucco C, Hough D, Thase ME, Shelton RC, Molero P. 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. American Journal of Psychiatry. 2019 Jun 1;176(6):428-438.
  62. 62. Swainson J, Thomas RK, Archer S, Chrenek C, MacKay MA, Baker G, Dursun S, Klassen LJ, Chokka P, Demas ML. Esketamine for treatment resistant depression. Expert review of neurotherapeutics. 2019 Oct 3;19(10):899-911.
  63. 63. Talbot J, Phillips JL, Blier P. Ketamine for chronic depression: Two cautionary tales. Journal of psychiatry & neuroscience: JPN. 2019 Nov;44(6):384.
  64. 64. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017 Mar 9;168(6):960-976.
  65. 65. Altamura C, Maes M, Dai J, Meltzer HY. Plasma concentrations of excitatory amino acids, serine, glycine, taurine and histidine in major depression. European Neuropsychopharmacology. 1995 Jan 1;5:71-75.
  66. 66. Küçükibrahimoğlu E, Saygın MZ, Çalışkan M, Kaplan OK, Ünsal C, Gören MZ. The change in plasma GABA, glutamine and glutamate levels in fluoxetine-or S-citalopram-treated female patients with major depression. European journal of clinical pharmacology. 2009 Jun;65(6):571-577.
  67. 67. Mitani H, Shirayama Y, Yamada T, Maeda K, Ashby Jr CR, Kawahara R. Correlation between plasma levels of glutamate, alanine and serine with severity of depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2006 Aug 30;30(6):1155-1158.
  68. 68. Krystal JH, Sanacora G, Duman RS. Rapid-acting glutamatergic antidepressants: The path to ketamine and beyond. Biological psychiatry. 2013 Jun 15;73(12):1133-1141.
  69. 69. Abdallah CG, Sanacora G, Duman RS, Krystal JH. Ketamine and rapid-acting antidepressants: A window into a new neurobiology for mood disorder therapeutics. Annual review of medicine. 2015 Jan 14;66:509-523.
  70. 70. Aleksandrova LR, Phillips AG, Wang YT. Antidepressant effects of ketamine and the roles of AMPA glutamate receptors and other mechanisms beyond NMDA receptor antagonism. Journal of psychiatry & neuroscience: JPN. 2017 Jul;42(4):222.
  71. 71. Strasburger SE, Bhimani PM, Kaabe JH, Krysiak JT, Nanchanatt DL, Nguyen TN, Pough KA, Prince TA, Ramsey NS, Savsani KH, Scandlen L. What is the mechanism of Ketamine's rapid-onset antidepressant effect? A concise overview of the surprisingly large number of possibilities. Journal of clinical pharmacy and therapeutics. 2017 Apr;42(2):147-154.
  72. 72. Moghaddam B, Adams B, Verma A, Daly D. 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. Journal of Neuroscience. 1997 Apr 15;17(8):2921-2927.
  73. 73. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, Kavalali ET, Monteggia LM. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011 Jul;475(7354):91-95.
  74. 74. Koike H, Iijima M, Chaki S. Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression. Behavioural brain research. 2011 Oct 10;224(1):107-111.
  75. 75. Maeng S, Zarate Jr CA, Du J, Schloesser RJ, McCammon J, Chen G, Manji HK. Cellular mechanisms underlying the antidepressant effects of ketamine: Role of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biological psychiatry. 2008 Feb 15;63(4):349-352.
  76. 76. El Iskandrani KS, Oosterhof CA, El Mansari M, Blier P. Impact of subanesthetic doses of ketamine on AMPA-mediated responses in rats: An in vivo electrophysiological study on monoaminergic and glutamatergic neurons. Journal of Psychopharmacology. 2015 Jul;29(7):792-801.
  77. 77. Björkholm C, Jardemark K, Schilström B, Svensson TH. Ketamine-like effects of a combination of olanzapine and fluoxetine on AMPA and NMDA receptor-mediated transmission in the medial prefrontal cortex of the rat. European Neuropsychopharmacology. 2015 Oct 1;25(10):1842-1847.
  78. 78. Nosyreva E, Szabla K, Autry AE, Ryazanov AG, Monteggia LM, Kavalali ET. Acute suppression of spontaneous neurotransmission drives synaptic potentiation. Journal of Neuroscience. 2013 Apr 17;33(16):6990-7002.
  79. 79. Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY, Aghajanian G, Duman RS. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010 Aug 20;329(5994):959-964.
  80. 80. Yang C, Hu YM, Zhou ZQ, Zhang GF, Yang JJ. Acute administration of ketamine in rats increases hippocampal BDNF and mTOR levels during forced swimming test. Upsala journal of medical sciences. 2013 Mar 1;118(1):3-8.
  81. 81. Yang C, Li WY, Yu HY, Gao ZQ, Liu XL, Zhou ZQ, Yang JJ. Tramadol pretreatment enhances ketamine-induced antidepressant effects and increases mammalian target of rapamycin in rat hippocampus and prefrontal cortex. Journal of Biomedicine and Biotechnology. 2012 Oct;2012.
  82. 82. Zhou W, Wang N, Yang C, Li XM, Zhou ZQ, Yang JJ. Ketamine-induced antidepressant effects are associated with AMPA receptors-mediated upregulation of mTOR and BDNF in rat hippocampus and prefrontal cortex. European Psychiatry. 2014 Sep;29(7):419-423.
  83. 83. Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, Li XY, Aghajanian G, Duman RS. Glutamate N-methyl-D-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biological psychiatry. 2011 Apr 15;69(8):754-761.
  84. 84. Liu RJ, Lee FS, Li XY, Bambico F, Duman RS, Aghajanian GK. Brain-derived neurotrophic factor Val66Met allele impairs basal and ketamine-stimulated synaptogenesis in prefrontal cortex. Biological psychiatry. 2012 Jun 1;71(11):996-1005.
  85. 85. Tornese P, Sala N, Bonini D, Bonifacino T, La Via L, Milanese M, Treccani G, Seguini M, Ieraci A, Mingardi J, Nyengaard JR. Chronic mild stress induces anhedonic behavior and changes in glutamate release, BDNF trafficking and dendrite morphology only in stress vulnerable rats. The rapid restorative action of ketamine. Neurobiology of stress. 2019 Feb 1;10:100160.
  86. 86. Dong C, Zhang JC, Yao W, Ren Q, Ma M, Yang C, Chaki S, Hashimoto K. Rapid and sustained antidepressant action of the mGlu2/3 receptor antagonist MGS0039 in the social defeat stress model: Comparison with ketamine. International Journal of Neuropsychopharmacology. 2017 Mar 1;20(3):228-236.
  87. 87. Laje G, Lally N, Mathews D, Brutsche N, Chemerinski A, Akula N, Kelmendi B, Simen A, McMahon FJ, Sanacora G, Zarate Jr C. Brain-derived neurotrophic factor Val66Met polymorphism and antidepressant efficacy of ketamine in depressed patients. Biological psychiatry. 2012 Dec 1;72(11):e27.
  88. 88. Chen MH, Lin WC, Wu HJ, Cheng CM, Li CT, Hong CJ, Tu PC, Bai YM, Tsai SJ, Su TP. Antisuicidal effect, BDNF Val66Met polymorphism, and low-dose ketamine infusion: Reanalysis of adjunctive ketamine study of Taiwanese patients with treatment-resistant depression (AKSTP-TRD). Journal of affective disorders. 2019 May 15;251:162-169.
  89. 89. Yang C, Zhou ZQ, Gao ZQ, Shi JY, Yang JJ. 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. Biological psychiatry. 2013 Jun 15;73(12):e35-e36.
  90. 90. Abdallah CG, Averill LA, Gueorguieva R, Goktas S, Purohit P, Ranganathan M, D’Souza DC, Formica R, Southwick SM, Duman RS, Sanacora G. 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. 2018 Jan 1:500959.
  91. 91. Ishiwata S, Hattori K, Sasayama D, Teraishi T, Miyakawa T, Yokota Y, Matsumura R, Nishikawa T, Kunugi H. Cerebrospinal fluid D-serine concentrations in major depressive disorder negatively correlate with depression severity. Journal of affective disorders. 2018 Jan 15;226:155-162.
  92. 92. Hashimoto K, Yoshida T, Ishikawa M, Fujita Y, Niitsu T, Nakazato M, Watanabe H, Sasaki T, Shiina A, Hashimoto T, Kanahara N. Increased serum levels of serine enantiomers in patients with depression. Actaneuropsychiatrica. 2016 Jun;28(3):173-178.
  93. 93. Malkesman O, Austin DR, Tragon T, Wang G, Rompala G, Hamidi AB, Cui Z, Young WS, Nakazawa K, Zarate Jr CA, Manji HK. Acute D-serine treatment produces antidepressant-like effects in rodents. International Journal of Neuropsychopharmacology. 2012 Sep 1;15(8):1135-1148.
  94. 94. Wei IH, Chen KT, Tsai MH, Wu CH, Lane HY, Huang CC. Acute amino acid D-serine administration, similar to ketamine, produces antidepressant-like effects through identical mechanisms. Journal of agricultural and food chemistry. 2017 Dec 13;65(49):10792-10803.
  95. 95. Otte DM, Barcena de Arellano ML, Bilkei-Gorzo A, Albayram Ö, Imbeault S, Jeung H, Alferink J, Zimmer A. Effects of chronic D-serine elevation on animal models of depression and anxiety-related behavior. PloS one. 2013 Jun 21;8(6):e67131.
  96. 96. Singh NS, Bernier M, Camandola S, Khadeer MA, Moaddel R, Mattson MP, Wainer IW. Enantioselective inhibition of d-serine transport by (S)-ketamine. British journal of pharmacology. 2015 Sep;172(18):4546-4559.
  97. 97. Singh NS, Rutkowska E, Plazinska A, Khadeer M, Moaddel R, Jozwiak K, Bernier M, Wainer IW. Ketamine metabolites enantioselectively decrease intracellular D-serine concentrations in PC-12 cells. PloS one. 2016 Apr 20;11(4):e0149499.
  98. 98. Moaddel R, Luckenbaugh DA, Xie Y, Villaseñor A, Brutsche NE, Machado-Vieira R, Ramamoorthy A, Lorenzo MP, Garcia A, Bernier M, Torjman MC. D-serine plasma concentration is a potential biomarker of (R, S)-ketamine antidepressant response in subjects with treatment-resistant depression. Psychopharmacology. 2015 Jan;232(2):399-409.
  99. 99. Hashimoto K. Blood D-serine levels as a predictive biomarker for the rapid antidepressant effects of the NMDA receptor antagonist ketamine. Psychopharmacology. 2014 Oct;231(20):4081-4082.
  100. 100. Williams NR, Heifets BD, Bentzley BS, Blasey C, Sudheimer KD, Hawkins J, Lyons DM, Schatzberg AF. Attenuation of antidepressant and antisuicidal effects of ketamine by opioid receptor antagonism. Molecular psychiatry. 2019 Dec;24(12):1779-1786.
  101. 101. Williams NR, Heifets BD, Blasey C, Sudheimer K, Pannu J, Pankow H, Hawkins J, Birnbaum J, Lyons DM, Rodriguez CI, Schatzberg AF. Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. American Journal of Psychiatry. 2018 Dec 1;175(12):1205-1215.
  102. 102. Yoon G, Petrakis IL, Krystal JH. Association of combined naltrexone and ketamine with depressive symptoms in a case series of patients with depression and alcohol use disorder. JAMA psychiatry. 2019 Mar 1;76(3):337-338.
  103. 103. Marton T, Barnes DE, Wallace A, Woolley JD. Concurrent use of buprenorphine, methadone, or naltrexone does not inhibit ketamine’s antidepressant activity. Biological psychiatry. 2019 Jun 15;85(12):e75-e76.
  104. 104. Molero P, Ramos-Quiroga JA, Martin-Santos R, Calvo-Sánchez E, Gutiérrez-Rojas L, Meana JJ. Antidepressant efficacy and tolerability of ketamine and esketamine: A critical review. CNS drugs. 2018 May;32(5):411-420.
  105. 105. Ho RC, Zhang MW. Ketamine as a rapid antidepressant: The debate and implications. BJPsych Advances. 2016 Jul;22(4):222-233.
  106. 106. Short B, Fong J, Galvez V, Shelker W, Loo CK. Side-effects associated with ketamine use in depression: A systematic review. The Lancet Psychiatry. 2018 Jan 1;5(1):65-78.
  107. 107. Schatzberg AF. A word to the wise about intranasal esketamine. American Journal of Psychiatry. 2019 Jun 1;176(6):422-424.
  108. 108. Sanacora G, Frye MA, McDonald W, Mathew SJ, Turner MS, Schatzberg AF, Summergrad P, Nemeroff CB, American Psychiatric Association. A consensus statement on the use of ketamine in the treatment of mood disorders. JAMA psychiatry. 2017 Apr 1;74(4):399-405.

Written By

Atamjit Singh and Preet Mohinder Singh Bedi

Submitted: 12 July 2021 Reviewed: 02 August 2021 Published: 10 September 2021