633
Views
17
CrossRef citations to date
0
Altmetric
Review

Beyond serotonin: newer antidepressants in the future

, &
Pages 777-790 | Received 24 Feb 2017, Accepted 08 Jun 2017, Published online: 19 Jun 2017

References

  • Undurraga J, Baldessarini RJ. Randomized, placebo-controlled trials of antidepressants for acute major depression: thirty-year meta-analytic review. Neuropsychopharmacology. 2012;37:851–864.
  • Tedeschini E, Levkovitz Y, Iovieno N, et al. Efficacy of antidepressants for late-life depression: a meta-analysis and meta-regression of placebo-controlled randomized trials. J Clin Psychiatry. 2011;72:1660–1668. .
  • Gaynes BN, Warden D, Trivedi MH, et al. What did STAR* D teach us? Results from a large-scale, practical, clinical trial for patients with depression. Psychiatr Services. 2009;60:1439–1445. .
  • Trivedi MH, Rush AJ, Wisniewski SR, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR* D: implications for clinical practice. Am J Psychiatry. 2006;163:28–40.
  • Souery D, Papakostas GI, Trivedi MH. Treatment-resistant depression. J Clin Psychiatry. 2006;67(Suppl 6):16–22.
  • Malhi GS, Byrow Y. Is treatment-resistant depression a useful concept? Evid Based Ment Health. 2016;19:1–3. .
  • Malhi GS, Parker GB, Crawford J, et al. Treatment-resistant depression: resistant to definition? Acta Psychiatr Scand. 2005;112:302–309.
  • Conway CR, George MS, Sackeim HA. Toward an evidence-based, operational definition of treatment-resistant depression: when enough is enough. JAMA Psychiatry. 2017;74:9-10.
  • Transcript on Treatment Resistant Depression. Centers for Medicare & Medicaid services; 2016. Available from: https://www.cms.gov/Regulations-and-Guidance/Guidance/FACA/downloads/id71d.pdf
  • Greenberg PE, Fournier AA, Sisitsky T, et al. The economic burden of adults with major depressive disorder in the United States (2005 and 2010). J Clin Psychiatry. 2015;76:155–162.
  • Olchanski N, McInnis Myers M, Halseth M, et al. The economic burden of treatment-resistant depression. Clin Ther. 2013;35:512–522.
  • Rush AJ, Trivedi MH, Stewart JW, et al. Combining medications to enhance depression outcomes (CO-MED): acute and long-term outcomes of a single-blind randomized study. Am J Psychiatry. 2011;168:689–701.
  • CERC-301. Cerecor; 2016. Available from: http://ir.cerecor.com/press-releases/detail/30/cerecor-reports-top-line-data-from-cerc-301-phase-2-study
  • Sanacora G, Treccani G, Popoli M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology. 2012;62:63–77.
  • Popoli M, Yan Z, McEwen BS, et al. The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Reviews Neurosci. 2012;13:22–37.
  • Musazzi L, Treccani G, Popoli M. Functional and structural remodeling of glutamate synapses in prefrontal and frontal cortex induced by behavioral stress. Front Psychiatry. 2015;6:60.
  • Gao SF, Bao AM. Corticotropin-releasing hormone, glutamate, and gamma-aminobutyric acid in depression. Neuroscientist. 2011;17:124–144.
  • Yuan TF, Hou G. The effects of stress on glutamatergic transmission in the brain. Mol Neurobiol. 2015;51:1139–1143.
  • Graybeal C, Kiselycznyk C, Holmes A. Stress-induced deficits in cognition and emotionality: a role of glutamate. Curr Top Behav Neurosci. 2012;12:189–207.
  • Musazzi L, Racagni G, Popoli M. Stress, glucocorticoids and glutamate release: effects of antidepressant drugs. Neurochem Int. 2011;59:138–149.
  • Douglas RJ, Martin KA. Mapping the matrix: the ways of neocortex. Neuron. 2007;56:226–238.
  • Frye MA, Tsai GE, Huggins T, et al. Low cerebrospinal fluid glutamate and glycine in refractory affective disorder. Biol Psychiatry. 2007;61:162–166.
  • Levine J, Panchalingam K, Rapoport A, et al. Increased cerebrospinal fluid glutamine levels in depressed patients. Biol Psychiatry. 2000;47:586–593.
  • Choudary PV, Molnar M, Evans SJ, et al. Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression. Proc Natl Acad Sci USA. 2005;102:15653–15658.
  • Machado-Vieira R, Manji HK, Zarate CA. The role of the tripartite glutamatergic synapse in the pathophysiology and therapeutics of mood disorders. Neuroscientist. 2009;15:525–539.
  • Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23:1–21.
  • Dang YH, Ma XC, Zhang JC, et al. Targeting of NMDA receptors in the treatment of major depression. Curr Pharm Des. 2014;20:5151–5159.
  • Hashimoto K. The role of glutamate on the action of antidepressants. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:1558–1568.
  • Schatzberg AF. Development of new psychopharmacological agents for depression and anxiety. Psychiatr Clin North Am. 2015;38:379–393.
  • Mathews DC, Henter ID, Zarate CA. Targeting the glutamatergic system to treat major depressive disorder: rationale and progress to date. Drugs. 2012;72:1313–1333.
  • Reus GZ, Abelaira HM, Tuon T, et al. Glutamatergic NMDA receptor as therapeutic target for depression. Adv Protein Chem Struct Biol. 2016;103:169–202.
  • Wang J, Jing L, Toledo-Salas JC, et al. Rapid-onset antidepressant efficacy of glutamatergic system modulators: the neural plasticity hypothesis of depression. Neurosci Bull. 2015;31:75–86.
  • Krystal JH, Sanacora G, Duman RS. Rapid-acting glutamatergic antidepressants: the path to ketamine and beyond. Biol Psychiatry. 2013;73:1133–1141.
  • Niciu MJ, Ionescu DF, Richards EM, et al. Glutamate and its receptors in the pathophysiology and treatment of major depressive disorder. J Neural Transm. 2014;121:907–924.
  • Chitty KM, Lagopoulos J, Lee RS, et al. A systematic review and meta-analysis of proton magnetic resonance spectroscopy and mismatch negativity in bipolar disorder. Eur Neuropsychopharmacol. 2013;23:1348–1363.
  • Gigante AD, Bond DJ, Lafer B, et al. Brain glutamate levels measured by magnetic resonance spectroscopy in patients with bipolar disorder: a meta-analysis. Bipolar Disord. 2012;14:478–487.
  • Arnone D, Mumuni AN, Jauhar S, et al. Indirect evidence of selective glial involvement in glutamate-based mechanisms of mood regulation in depression: meta-analysis of absolute prefrontal neuro-metabolic concentrations. Eur Neuropsychopharmacol. 2015;25:1109–1117.
  • Lee HK, Kirkwood A. AMPA receptor regulation during synaptic plasticity in hippocampus and neocortex. Semin Cell Dev Biol. 2011;22:514–520.
  • Musazzi L, Treccani G, Mallei A, et al. The action of antidepressants on the glutamate system: regulation of glutamate release and glutamate receptors. Biol Psychiatry. 2013;73:1180–1188.
  • Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol. 2010;50:295–322.
  • Niciu MJ, Ionescu DF, Mathews DC, et al. Second messenger/signal transduction pathways in major mood disorders: moving from membrane to mechanism of action, part I: major depressive disorder. CNS Spectrums. 2013;18:231–241.
  • Monaghan DT, Jane DE. Pharmacology of NMDA receptors. In: Van Dongen AM, editor. Biology of the NMDA receptor. Boca Raton (FL): Frontiers in Neuroscience; 2009.
  • Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14:383–400.
  • Duman RS. Neurobiology of stress, depression, and rapid acting antidepressants: remodeling synaptic connections. Depress Anxiety. 2014;31:291–296.
  • Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010;11:682–696.
  • Chong C, Schug SA, Page-Sharp M, et al. Development of a sublingual/oral formulation of ketamine for use in neuropathic pain: preliminary findings from a three-way randomized, crossover study. Clin Drug Investig. 2009;29:317–324.
  • Coyle CM, Laws KR. The use of ketamine as an antidepressant: a systematic review and meta-analysis. Hum Psychopharmacol. 2015;30:152–163.
  • Caddy C, Giaroli G, White TP, et al. Ketamine as the prototype glutamatergic antidepressant: pharmacodynamic actions, and a systematic review and meta-analysis of efficacy. Ther Advances Psychopharmacol. 2014;4:75–99.
  • Fond G, Loundou A, Rabu C, et al. Ketamine administration in depressive disorders: a systematic review and meta-analysis. Psychopharmacology. 2014;231:3663–3676.
  • Singh JB, Fedgchin M, Daly EJ, et al. A double-blind, randomized, placebo-controlled, dose-frequency study of intravenous ketamine in patients with treatment-resistant depression. Am J Psychiatry. 2016;173:816–826.
  • Niesters M, Martini C, Dahan A. Ketamine for chronic pain: risks and benefits. Br J Clin Pharmacol. 2014;77:357–367.
  • Newport DJ, Carpenter LL, McDonald WM, et al. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172:950–966.
  • McGirr A, Berlim MT, Bond DJ, 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.
  • Wang X, Chen Y, Zhou X, et al. Effects of propofol and ketamine as combined anesthesia for electroconvulsive therapy in patients with depressive disorder. J Ect. 2012;28:128–132.
  • Loo CK, Katalinic N, Garfield JB, et al. Neuropsychological and mood effects of ketamine in electroconvulsive therapy: a randomised controlled trial. J Affect Disord. 2012;142:233–240.
  • Yoosefi A, Sepehri AS, Kargar M, et al. Comparing effects of ketamine and thiopental administration during electroconvulsive therapy in patients with major depressive disorder: a randomized, double-blind study. J Ect. 2014;30:15–21.
  • Abdallah CG, Fasula M, Kelmendi B, et al. Rapid antidepressant effect of ketamine in the electroconvulsive therapy setting. J Ect. 2012;28:157–161.
  • Jarventausta K, Chrapek W, Kampman O, et al. Effects of S-ketamine as an anesthetic adjuvant to propofol on treatment response to electroconvulsive therapy in treatment-resistant depression: a randomized pilot study. J Ect. 2013;29:158–161.
  • Abdallah CG, Adams TG, Kelmendi B, et al. Ketamine’s mechanism of action: a path to rapid-acting antidepressants. Depress Anxiety. 2016;33:689–697.
  • Abdallah CG, Averill LA, Krystal JH. Ketamine as a promising prototype for a new generation of rapid-acting antidepressants. Ann NY Acad Sci. 2015;1344:66–77.
  • Lepack AE, Fuchikami M, Dwyer JM, et al. BDNF release is required for the behavioral actions of ketamine. Int J Neuropsychopharmacol. 2014;18.
  • Autry AE, Adachi M, Nosyreva E, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011;475:91–95. .
  • Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329:959–964. .
  • Murrough JW, Collins KA, Fields J, et al. Regulation of neural responses to emotion perception by ketamine in individuals with treatment-resistant major depressive disorder. Transl Psychiatry. 2015;5:e509.
  • Salvadore G, Cornwell BR, Colon-Rosario V, et al. Increased anterior cingulate cortical activity in response to fearful faces: a neurophysiological biomarker that predicts rapid antidepressant response to ketamine. Biol Psychiatry. 2009;65:289–295.
  • Salvadore G, Cornwell BR, Sambataro F, et al. Anterior cingulate desynchronization and functional connectivity with the amygdala during a working memory task predict rapid antidepressant response to ketamine. Neuropsychopharmacology. 2010;35:1415–1422.
  • Di Lazzaro V, Oliviero A, Profice P, et al. Ketamine increases human motor cortex excitability to transcranial magnetic stimulation. J Physiol. 2003;547:485–496.
  • Duncan WC, Sarasso S, Ferrarelli F, et al. Concomitant BDNF and sleep slow wave changes indicate ketamine-induced plasticity in major depressive disorder. Int J Neuropsychopharmacol. 2013;16:301–311.
  • Bowdle TA, Radant AD, Cowley DS, et al. Psychedelic effects of ketamine in healthy volunteers: relationship to steady-state plasma concentrations. Anesthesiology. 1998;88:82–88.
  • Pomarol-Clotet E, Honey GD, Murray GK, et al. Psychological effects of ketamine in healthy volunteers. Phenomenological study. Br J Psychiatry. 2006;189:173–179.
  • Cvrcek P. Side effects of ketamine in the long-term treatment of neuropathic pain. Pain Med. 2008;9:253–257.
  • Morgan CJ, Curran HV. Acute and chronic effects of ketamine upon human memory: a review. Psychopharmacology (Berl). 2006;188:408–424.
  • Sigtermans M, Dahan A, Mooren R, et al. S(+)-ketamine effect on experimental pain and cardiac output: a population pharmacokinetic-pharmacodynamic modeling study in healthy volunteers. Anesthesiology. 2009;111:892–903. .
  • Zhu W, Ding Z, Zhang Y, et al. Risks associated with misuse of ketamine as a rapid-acting antidepressant. Neurosci Bull. 2016;32:557–564.
  • Middela S, Pearce I. Ketamine-induced vesicopathy: a literature review. Int J Clin Pract. 2011;65:27–30.
  • Nishimura M, Sato K. Ketamine stereoselectively inhibits rat dopamine transporter. Neurosci Lett. 1999;274:131–134.
  • Hashimoto K, Kakiuchi T, Ohba H, et al. Reduction of dopamine D2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys. Eur Arch Psychiatry Clin Neurosci. 2017;267:173-176.
  • Vollenweider FX, Leenders KL, Oye I, et al. Differential psychopathology and patterns of cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using positron emission tomography (PET). Eur Neuropsychopharmacol. 1997;7:25–38.
  • Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory perception: evidence for a role of N-methyl-D-aspartate receptors. J Pharmacol Exp Ther. 1992;260:1209–1213.
  • Kohrs R, Durieux ME. Ketamine: teaching an old drug new tricks. Anesth Analg. 1998;87:1186–1193.
  • Muller J, Pentyala S, Dilger J, et al. Ketamine enantiomers in the rapid and sustained antidepressant effects. Ther Advances Psychopharmacol. 2016;6:185–192.
  • Singh JB, Fedgchin M, Daly E, et al. Intravenous esketamine in adult treatment-resistant depression: a double-blind, double-randomization, placebo-controlled study. Biol Psychiatry. 2015;80:424-431.
  • Yang C, Shirayama Y, Zhang JC, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632.
  • Esketamine Breakthrough Designation. Janssen; 2016. Available from: http://www.jnj.com/news/all/Esketamine-Receives-Breakthrough-Therapy-Designation-from-US-Food-and-Drug-Administration-for-Major-Depressive-Disorder-with-Imminent-Risk-for-Suicide.
  • Bui KH, Zhou D, Agbo F, et al. Effect of multiple intravenous doses of lanicemine (AZD6765) on the pharmacokinetics of midazolam in healthy subjects. J Clin Pharmacol. 2015;55:1024–1030.
  • Downey D, Dutta A, McKie S, et al. Comparing the actions of lanicemine and ketamine in depression: key role of the anterior cingulate. Eur Neuropsychopharmacol. 2016;26:994–1003.
  • Guo J, Zhou D, Grimm SW, et al. Pharmacokinetics, metabolism and excretion of [(14)C]-lanicemine (AZD6765), a novel low-trapping N-methyl-d-aspartic acid receptor channel blocker, in healthy subjects. Xenobiotica. 2015;45:244–255.
  • Sanacora G, Smith MA, Pathak S, et al. Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects. Mol Psychiatry. 2014;19:978–985.
  • AZD6423. Astrazeneca; 2014. Available from: https://neurogram.wordpress.com/2014/02/06/astrazeneca-good-news-bad-news/
  • Graef JD, Newberry K, Newton A, et al. Effect of acute NR2B antagonist treatment on long-term potentiation in the rat hippocampus. Brain Res. 2015;1609:31–39.
  • Preskorn SH, Baker B, Kolluri S, et al. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2008;28:631–637.
  • Hashimoto K. Comments on an innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101606 in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2009;29:411–412; author reply 2.
  • Mott DD, Doherty JJ, Zhang S, et al. Phenylethanolamines inhibit NMDA receptors by enhancing proton inhibition. Nat Neurosci. 1998;1:659–667.
  • Ibrahim L, Diaz Granados N, Jolkovsky L, et al. A randomized, placebo-controlled, crossover pilot trial of the oral selective NR2B antagonist MK-0657 in patients with treatment-resistant major depressive disorder. J Clin Psychopharmacol. 2012;32:551–557.
  • Garner R, Gopalakrishnan S, McCauley JA, et al. Preclinical pharmacology and pharmacokinetics of CERC-301, a GluN2B-selective N-methyl-D-aspartate receptor antagonist. Pharmacol Res Perspect. 2015;3:e00198.
  • Stroebel D, Buhl DL, Knafels JD, et al. A novel binding mode reveals two distinct classes of NMDA receptor GluN2B-selective antagonists. Mol Pharmacol. 2016;89:541–551.
  • EVT-101. Evotec; 2009. Available from: https://www.evotec.com/uploads/cms_article/1520/Evotec_and_Roche_to_Develop_EVT_101.pdf
  • Heresco-Levy U, Gelfin G, Bloch B, et al. A randomized add-on trial of high-dose D-cycloserine for treatment-resistant depression. Int J Neuropsychopharmacol. 2013;16:501–506.
  • Heresco-Levy U, Javitt DC, Gelfin Y, et al. Controlled trial of D-cycloserine adjuvant therapy for treatment-resistant major depressive disorder. J Affect Disord. 2006;93:239–243.
  • Zhang XL, Sullivan JA, Moskal JR, et al. A NMDA receptor glycine site partial agonist, GLYX-13, simultaneously enhances LTP and reduces LTD at Schaffer collateral-CA1 synapses in hippocampus. Neuropharmacology. 2008;55:1238–1250.
  • Burgdorf J, Zhang XL, Weiss C, et al. The N-methyl-D-aspartate receptor modulator GLYX-13 enhances learning and memory, in young adult and learning impaired aging rats. Neurobiol Aging. 2011;32:698–706.
  • Preskorn S, Macaluso M, Mehra DO, et al. Randomized proof of concept trial of GLYX-13, an N-methyl-D-aspartate receptor glycine site partial agonist, in major depressive disorder nonresponsive to a previous antidepressant agent. J Psychiatr Pract. 2015;21:140–149.
  • Burch R. Phase 2 data for GLYX13. 2012. In: Proceedings of the 51st American College of Neuropsychopharmacology (ACNP) Conference; 2012 Dec 2-6; Hollywood, FL. Publisher: American College of Neuropsychopharmacology.
  • Fuxe K, Borroto-Escuela DO. Basimglurant for treatment of major depressive disorder: a novel negative allosteric modulator of metabotropic glutamate receptor 5. Expert Opin Investig Drugs. 2015;24:1247–1260.
  • Jaeschke G, Kolczewski S, Spooren W, et al. Metabotropic glutamate receptor 5 negative allosteric modulators: discovery of 2-chloro-4-[1-(4-fluorophenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]pyridine (basimglurant, RO4917523), a promising novel medicine for psychiatric diseases. J Med Chem. 2015;58:1358–1371.
  • Lindemann L, Porter RH, Scharf SH, et al. Pharmacology of basimglurant (RO4917523, RG7090), a unique metabotropic glutamate receptor 5 negative allosteric modulator in clinical development for depression. J Pharmacol Exp Ther. 2015;353:213–233.
  • Quiroz JA, Tamburri P, Deptula D, et al. Efficacy and safety of basimglurant as adjunctive therapy for major depression: a randomized clinical trial. JAMA Psychiatry. 2016;73:675–684.
  • Garay RP, Citrome L, Grossberg GT, et al. Investigational drugs for treating agitation in persons with dementia. Expert Opin Investig Drugs. 2016;25:973–983.
  • Lauterbach EC. Treatment resistant depression with loss of antidepressant response: rapid-acting antidepressant action of dextromethorphan, a possible treatment bridging molecule. Psychopharmacol Bull. 2016;46:53–58.
  • Zanos P, Piantadosi SC, Wu HQ, et al. The prodrug 4-chlorokynurenine causes ketamine-like antidepressant effects, but not side effects, by NMDA/GlycineB-site inhibition. J Pharmacol Exp Ther. 2015;355:76–85.
  • Liu BB, Luo L, Liu XL, et al. 7-Chlorokynurenic acid (7-CTKA) produces rapid antidepressant-like effects: through regulating hippocampal microRNA expressions involved in TrkB-ERK/Akt signaling pathways in mice exposed to chronic unpredictable mild stress. Psychopharmacology (Berl). 2015;232:541–550.
  • AV-101. Vistagen; 2017. Available from: http://www.vistagen.com/pipeline/av-101
  • Ehrich E, Turncliff R, Du Y, et al. Evaluation of opioid modulation in major depressive disorder. Neuropsychopharmacology. 2015;40:1448–1455. .
  • Kennedy SE, Koeppe RA, Young EA, et al. Dysregulation of endogenous opioid emotion regulation circuitry in major depression in women. Arch Gen Psychiatry. 2006;63:1199–1208.
  • Carlezon WA Jr., Beguin C, Knoll AT, et al. Kappa-opioid ligands in the study and treatment of mood disorders. Pharmacol Ther. 2009;123:334–343.
  • Negus SS, Morrissey EM, Rosenberg M, et al. Effects of kappa opioids in an assay of pain-depressed intracranial self-stimulation in rats. Psychopharmacology (Berl). 2010;210:149–159.
  • Tomasiewicz HC, Todtenkopf MS, Chartoff EH, et al. The kappa-opioid agonist U69593 blocks cocaine-induced enhancement of brain stimulation reward. Biol Psychiatry. 2008;64:982–988.
  • Carlezon WA Jr., Krystal AD. Kappa-opioid antagonists for psychiatric disorders: from bench to clinical trials. Depress Anxiety. 2016;33:895–906.
  • Carlezon WA Jr., Thome J, Olson VG, et al. Regulation of cocaine reward by CREB. Science. 1998;282:2272–2275.
  • Carroll FI, Carlezon WA Jr. Development of kappa opioid receptor antagonists. J Med Chem. 2013;56:2178–2195. .
  • Yovell Y, Bar G, Mashiah M, et al. Ultra-low-dose buprenorphine as a time-limited treatment for severe suicidal ideation: a randomized controlled trial. Am J Psychiatry. 2016;173:491–498.
  • Wentland MP, Lou R, Lu Q, et al. Syntheses and opioid receptor binding properties of carboxamido-substituted opioids. Bioorg Med Chem Lett. 2009;19:203–208.
  • Wentland MP, Lu Q, Lou R, et al. Synthesis and opioid receptor binding properties of a highly potent 4-hydroxy analogue of naltrexone. Bioorg Med Chem Lett. 2005;15:2107–2110.
  • Fava M, Memisoglu A, Thase ME, et al. Opioid modulation with buprenorphine/samidorphan as adjunctive treatment for inadequate response to antidepressants: a randomized double-blind placebo-controlled trial. Am J Psychiatry. 2016;173:499–508.
  • Lowe SL, Wong CJ, Witcher J, et al. Safety, tolerability, and pharmacokinetic evaluation of single- and multiple-ascending doses of a novel kappa opioid receptor antagonist LY2456302 and drug interaction with ethanol in healthy subjects. J Clin Pharmacol. 2014;54:968–978.
  • Janowsky DS, el-Yousef MK, Davis JM, et al. Cholinergic reversal of manic symptoms. Lancet. 1972;1:1236–1237.
  • Risch SC, Kalin NH, Janowsky DS. Cholinergic challenges in affective illness: behavioral and neuroendocrine correlates. J Clin Psychopharmacol. 1981;1:186–192.
  • Carroll BJ, Frazer A, Schless A, et al. Cholinergic reversal of manic symptoms. Lancet. 1973;1:427–428.
  • Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial. Arch Gen Psychiatry. 2006;63:1121–1129.
  • Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010;67:432–438.
  • Furey ML, Khanna A, Hoffman EM, et al. Scopolamine produces larger antidepressant and antianxiety effects in women than in men. Neuropsychopharmacology. 2010;35:2479–2488.
  • Drevets WC, Zarate CA Jr., Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73:1156–1163.
  • Voleti B, Navarria A, Liu RJ, et al. Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry. 2013;74:742–749.
  • Shelton RC, Pencina MJ, Barrentine LW, et al. Association of obesity and inflammatory marker levels on treatment outcome: results from a double-blind, randomized study of adjunctive L-methylfolate calcium in patients with MDD who are inadequate responders to SSRIs. J Clin Psychiatry. 2015;76:1635–1641.
  • Kim YK, Won E. The influence of stress on neuroinflammation and alterations in brain structure and function in major depressive disorder. Behav Brain Res. 2017;329:6–11.
  • Sharma A. Systems genomics support for immune and inflammation hypothesis of depression. Curr Neuropharmacol. 2016;14:749–758.
  • Spalletta G, Bossu P, Ciaramella A, et al. The etiology of poststroke depression: a review of the literature and a new hypothesis involving inflammatory cytokines. Mol Psychiatry. 2006;11:984–991.
  • Maes M, Yirmyia R, Noraberg J, et al. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression. Metab Brain Dis. 2009;24:27–53.
  • Liu Y, Ho RC, Mak A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. J Affect Disord. 2012;139:230–239.
  • Fond G, Hamdani N, Kapczinski F, et al. Effectiveness and tolerance of anti-inflammatory drugs’ add-on therapy in major mental disorders: a systematic qualitative review. Acta Psychiatr Scand. 2014;129:163–179.
  • Raison CL, Rutherford RE, Woolwine BJ, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry. 2013;70:31–41.
  • Miller LL, Leitl MD, Banks ML, et al. Effects of the triple monoamine uptake inhibitor amitifadine on pain-related depression of behavior and mesolimbic dopamine release in rats. Pain. 2015;156:175–184.
  • Marks DM, Abramowitz JS, Spielmans GI. Concerns about data reporting and interpretation in “efficacy and tolerability of the novel triple reuptake inhibitor amitifadine in the treatment of patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial”. J Psychiatr Res. 2012;46:692–693; author reply 4-5.
  • Tran P, Skolnick P, Czobor P, et al. Efficacy and tolerability of the novel triple reuptake inhibitor amitifadine in the treatment of patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. J Psychiatr Res. 2012;46:64–71.
  • Femenia T, Magara S, DuPont CM, et al. Hippocampal-dependent antidepressant action of the H3 receptor antagonist clobenpropit in a rat model of depression. Int J Neuropsychopharmacol. 2015;18.
  • Tiligada E, Kyriakidis K, Chazot PL, et al. Histamine pharmacology and new CNS drug targets. CNS Neurosci Ther. 2011;17:620–628.
  • Passani MB, Blandina P. Histamine receptors in the CNS as targets for therapeutic intervention. Trends Pharmacol Sci. 2011;32:242–249.
  • Ring RM, Regan CM. Captodiamine, a putative antidepressant, enhances hypothalamic BDNF expression in vivo by synergistic 5-HT2c receptor antagonism and sigma-1 receptor agonism. J Psychopharmacol. 2013;27:930–939.
  • Fava M, Johe K, Ereshefsky L, et al. A phase 1B, randomized, double blind, placebo controlled, multiple-dose escalation study of NSI-189 phosphate, a neurogenic compound, in depressed patients. Molecular Psychiatry. 2016;21:1372-1380.
  • Fava M, Johe K, Ereshefsky L, et al. A phase 1B, randomized, double blind, placebo controlled, multiple-dose escalation study of NSI-189 phosphate, a neurogenic compound, in depressed patients. Molecular Psychiatry. 2016;21:1483-1484.
  • Dos Santos RG, Osorio FL, Crippa JA, et al. Antidepressive, anxiolytic, and antiaddictive effects of ayahuasca, psilocybin and lysergic acid diethylamide (LSD): a systematic review of clinical trials published in the last 25 years. Ther Adv Psychopharmacol. 2016;6:193–213. .
  • Carhart-Harris RL, Goodwin GM. The therapeutic potential of psychedelic drugs: past, present, and future. Neuropsychopharmacology. 2017.
  • Carhart-Harris RL, Bolstridge M, Rucker J, et al. Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. Lancet Psychiatry. 2016;3:619–627.
  • Griffiths RR, Johnson MW, Carducci MA, et al. Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: a randomized double-blind trial. J Psychopharmacol. 2016;30:1181–1197.
  • Osorio Fde L, Sanches RF, Macedo LR, et al. Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: a preliminary report. Rev Bras Psiquiatr. 2015;37:13–20.
  • Sanches RF, de Lima Osorio F, Dos Santos RG, et al. Antidepressant effects of a single dose of Ayahuasca in patients with recurrent depression: a SPECT study. J Clin Psychopharmacol. 2016;36:77–81.
  • dos Santos RG, Balthazar FM, Bouso JC, et al. The current state of research on Ayahuasca: a systematic review of human studies assessing psychiatric symptoms, neuropsychological functioning, and neuroimaging. J Psychopharmacol. 2016;30:1230–1247.
  • McCorvy JD, Olsen RH, Roth BL. Psilocybin for depression and anxiety associated with life-threatening illnesses. J Psychopharmacol. 2016;30:1209–1210.
  • Kellner CH, Fink M. Electroconvulsive therapy versus pharmacotherapy for bipolar depression. Am J Psychiatry. 2015;172:295.
  • Schoeyen HK, Kessler U, Andreassen OA, et al. Treatment-resistant bipolar depression: a randomized controlled trial of electroconvulsive therapy versus algorithm-based pharmacological treatment. Am J Psychiatry. 2015;172:41–51.
  • Kellner CH, Husain MM, Knapp RG, et al. A novel strategy for continuation ECT in geriatric depression: phase 2 of the PRIDE study. Am J Psychiatry. 2016;173:1110-1118.
  • Kellner CH, Husain MM, Knapp RG, et al. Right unilateral ultrabrief pulse ECT in geriatric depression: phase 1 of the PRIDE study. Am J Psychiatry. 2016;173:1101-1109.
  • Milev RV, Giacobbe P, Kennedy SH, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 4. neurostimulation treatments. Can J Psychiatry Revue Canadienne De Psychiatrie. 2016;61:561–575.
  • Dell’Osso B, Oldani L, Camuri G, et al. Augmentative repetitive Transcranial Magnetic Stimulation (rTMS) in the acute treatment of poor responder depressed patients: a comparison study between high and low frequency stimulation. Eur Psychiatry. 2015;30:271–276.
  • Dunner DL, Aaronson ST, Sackeim HA, et al. A multisite, naturalistic, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a 1-year follow-up period. J Clin Psychiatry. 2014;75:1394–1401.
  • Kedzior KK, Gellersen HM, Brachetti AK, et al. Deep transcranial magnetic stimulation (DTMS) in the treatment of major depression: an exploratory systematic review and meta-analysis. J Affect Disord. 2015;187:73–83.
  • Levkovitz Y, Isserles M, Padberg F, et al. Efficacy and safety of deep transcranial magnetic stimulation for major depression: a prospective multicenter randomized controlled trial. World Psychiatry. 2015;14:64–73.
  • Leuchter AF, Cook IA, Feifel D, et al. Efficacy and safety of low-field synchronized Transcranial Magnetic Stimulation (sTMS) for treatment of major depression. Brain Stimul. 2015;8:787–794.
  • Rohan ML, Yamamoto RT, Ravichandran CT, et al. Rapid mood-elevating effects of low field magnetic stimulation in depression. Biol Psychiatry. 2014;76:186–193.
  • Yang C, Zhou ZQ, Gao ZQ, et al. Acute increases in plasma mammalian target of rapamycin, glycogen synthase kinase-3beta, and eukaryotic elongation factor 2 phosphorylation after ketamine treatment in three depressed patients. Biol Psychiatry. 2013;73:e35–e36.
  • Slyepchenko A, Carvalho AF, Cha DS, et al. Gut emotions - mechanisms of action of probiotics as novel therapeutic targets for depression and anxiety disorders. CNS Neurol Disord Drug Targets. 2014;13:1770–1786.
  • Garay RP, Zarate CA Jr., Charpeaud T, et al. Investigational drugs in recent clinical trials for treatment-resistant depression. Expert Rev Neurother. 2017;17:593–609. .
  • Bunney BG, Li JZ, Walsh DM, et al. Circadian dysregulation of clock genes: clues to rapid treatments in major depressive disorder. Molecular Psychiatry. 2015;20:48–55.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.