352
Views
0
CrossRef citations to date
0
Altmetric
Review

Clinical pharmacological innovation in the treatment of depression

, & ORCID Icon
Pages 349-362 | Received 16 Feb 2023, Accepted 30 Mar 2023, Published online: 06 Apr 2023

References

  • https://www.nimh.nih.gov/health/statistics/major-depression/
  • American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed., text rev. Washington, DC: American Psychiatric Publishing, Inc; 2022.
  • https://pubmed.ncbi.nlm.nih.gov/
  • https://clinicaltrials.gov
  • Witkin JM, Martin AE, Smith JL. Muscarinic cholinergic receptor antagonism: one doorway to rapid relief from major depressive disorder. In: Duncan L, editor. Advances in Health and Disease. Vol. 61. Hauppauge (NY): Nova Science Publishers, Inc; 2023. p. 210–223.
  • Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR_D report. Amer J Psychiat. 2006;163:1905–1917.
  • Katz MM, Koslow SH, Frazer A. Onset of antidepressant activity: reexamining the structure of depression and multiple actions of drugs. Depress Anxiety. 1996;4(6):257–267.
  • Katz MM, Tekell JL, Bowden CL, et al. Onset and early behavioral effects of pharmacologically different antidepressants and placebo in depression. Neuropsychopharmacology. 2004;29:566–579.
  • 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 Serv. 2009;60(11):1439–1445. DOI:10.1176/ps.2009.60.11.1439.
  • Locher C, Koechlin H, Zion SR, et al. Efficacy and safety of selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors, and placebo for common psychiatric disorders among children and adolescents: a systematic review and meta-analysis. JAMA Psychiatry. 2017;74(10):1011–1020. DOI:10.1001/jamapsychiatry.2017.2432.
  • Fava M, Ball S, Nelson JC, et al. Clinical relevance of fatigue as a residual symptom in major depressive disorder. Depress Anxiety. 2014;31(3):250–257. DOI:10.1002/da.22199.
  • McIntyre RS, Harrison J, Loft H, et al. The effects of vortioxetine on cognitive function in patients with major depressive disorder: a meta-analysis of three randomized controlled trials. Inter J Neuropsychopharmacol. 2016;19(10): pyw055. DOI:10.1093/ijnp/pyw055.
  • Ferguson JM. SSRI antidepressant medications: adverse effects and tolerability. Primary Care Companion to the J Clin Psychiatry. 2001;3(1):22–27.
  • Khawam EA, Laurencic G, DA M Jr. Side effects of antidepressants: an overview. Clev Clin J Med. 2006;73(4): 351-3, 356–61. DOI:10.3949/ccjm.73.4.351.
  • Solmi M, Miola A, Croatto G, et al. How can we improve antidepressant adherence in the management of depression? A targeted review and 10 clinical recommendations. Braz J Psychiatry. 2021;43(2):189–202. DOI:10.1590/1516-4446-2020-0935.
  • Dunlop K, Talishinsky A, Liston C. Intrinsic brain network biomarkers of antidepressant response: a review. Curr Psychiat Rep. 2019;21(9):87.
  • Voegeli G, Cléry-Melin ML, Ramoz N, et al. Progress in elucidating biomarkers of antidepressant pharmacological treatment response: a systematic review and meta-analysis of the last 15 years. Drugs. 2017;77(18):1967–1986. DOI:10.1007/s40265-017-0819-9.
  • Targum SD, Schappi J, Koutsouris A, et al. A novel peripheral biomarker for depression and antidepressant response. Mol Psychiatry. 2022;27(3):1640–1646. DOI:10.1038/s41380-021-01399-1.
  • Costi S, Evers A, Jha MK, et al. A randomized pilot study of the prophylactic effect of ketamine on laboratory-induced stress in healthy adults. Neurobiol Stress. 2023;22:100505.
  • Czarny P, Białek K, Ziółkowska S, et al. The importance of epigenetics in diagnostics and treatment of major depressive disorder. J Person Med. 2021;11(3):167. DOI:10.3390/jpm11030167.
  • Glennon RA, Iversen L. Antidepressants. In: editors, Abraham D, and Myers M. Burger’s medicinal chemistry, drug discovery and development. 8th ed. Hoboken (NJ): John Wiley and Sons, Inc; 2021. p. 1–72.
  • De Diego-Adeliño J, Crespo JM, Mora F, et al. Vortioxetine in major depressive disorder: from mechanisms of action to clinical studies. An updated review. Exp Opin Drug Safety. 2022;21(5):673–690. DOI:10.1080/14740338.2022.2019705.
  • Bleacher H, Koerperich L, Malam N, et al. Are antipsychotics effective adjunctive Tx for patients with moderate-to-severe depression? J Fam Practice. 2022;71(6):E13–15. DOI:10.12788/jfp.0445.
  • Han C, Wang SM, Kato M, et al. Second-generation antipsychotics in the treatment of major depressive disorder: current evidence. Expert Rev Neurother. 2013;13(7):851–870. DOI:10.1586/14737175.2013.811901.
  • Ruberto VL, Jha MK, Murrough JW. Pharmacological treatments for patients with treatment-resistant depression. Pharmaceuticals. 2020;13(6):116.
  • Rogóż Z. Combined treatment with atypical antipsychotics and antidepressants in treatment-resistant depression: preclinical and clinical efficacy. Pharmacol Rep. 2013;65(6):1535–1544.
  • Shelton RC, Papakostas GI. Augmentation of antidepressants with atypical antipsychotics for treatment-resistant major depressive disorder. Acta Psychiatr Scand. 2008;117(4):253–259.
  • Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharmacol. 1990;185(1):1–10. DOI:10.1016/0014-2999(90)90204-J.
  • Skolnick P, Popik P, Trullas R. Glutamate-based antidepressants: 20 years on. Trends Pharmacol Sci. 2009;30(11):563–569.
  • Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47(4):351–354. DOI:10.1016/S0006-3223(99)00230-9.
  • Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856–864. DOI:10.1001/archpsyc.63.8.856.
  • https://www.spravatohcp.com
  • Alnefeesi Y, Chen-Li D, Krane E, et al. Real-world effectiveness of ketamine in treatment-resistant depression: a systematic review & meta-analysis. J Psychiatr Res. 2022;151:693–709.
  • Fornaro M, De Berardis D, Anastasia A, et al. Novel psychopharmacology for depressive disorders. Adv Exp Med Biol. 2021;1305:449–461.
  • Jawad MY, Di Vincenzo JD, Badulescu S, et al. The therapeutic role of ketamine and esketamine in treating psychopathological domains of depression. Neuropharmacology. 2023;223:109299.
  • Price RB, Kissel N, Baumeister A. International pooled patient-level meta-analysis of ketamine infusion for depression: in search of clinical moderators. Mol Psychiat. 2022;27(12):5096–5112. DOI:10.1038/s41380-022-01757-7.
  • Smith-Apeldoorn SY, Veraart JK, Spijker J, et al. Maintenance ketamine treatment for depression: a systematic review of efficacy, safety, and tolerability. Lancet Psychiat. 2022;9(11):907–921. DOI:10.1016/S2215-0366(22)00317-0.
  • Witkin JM, Martin AE, Golani LK, et al. Rapid-acting antidepressants. Adv Pharmacol. 2019;86:47–96.
  • Yavi M, Lee H, Henter ID, et al. Ketamine treatment for depression: a review. Discover Mental Health. 2022;2(1):9. DOI:10.1007/s44192-022-00012-3.
  • Zanos P, Moaddel R, Morris PJ, et al. Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharmacol Rev. 2018;70(3):621–660. DOI:10.1124/pr.117.015198.
  • Bahji A, Zarate CA, Vazquez GH. Efficacy and safety of racemic ketamine and esketamine for depression: a systematic review and meta-analysis. Exp Opin Drug Safety. 2022;21(6):853–866. DOI:10.1080/14740338.2022.2047928.
  • De Berardis D, Tomasetti C, Pompili M, et al. An Update on glutamatergic system in suicidal depression and on the role of esketamine. Curr Top Med Chem. 2020;20(7):554–584. PMID: 32003691. DOI:10.2174/1568026620666200131100316.
  • Liu P, Zhang SS, Liang Y, et al. Efficacy and safety of esketamine combined with antidepressants for treatment-resistant depression: a meta-analysis. Neuropsychiatr Dis Treat. 2022;18:2855–2865.
  • Psiuk D, Nowak EM, Dycha N, et al. Esketamine and psilocybin-the comparison of two mind-altering agents in depression treatment: systematic review. Int J Mol Sci. 2022;23(19):11450. DOI:10.3390/ijms231911450.
  • Pilc A, Machaczka A, Kawalec P, et al. Where do we go next in antidepressant drug discovery? A new generation of antidepressants: a pivotal role of AMPA receptor potentiation and mGlu2/3 receptor antagonism. Exp Opin Drug Discovery. 2022;17(10):1131–1146. DOI:10.1080/17460441.2022.2111415.
  • Hess EM, Riggs LM, Michaelides M, et al. Mechanisms of ketamine and its metabolites as antidepressants. Biochem Pharmacol. 2022;197:114892.
  • Lumsden EW, Troppoli TA, Myers SJ, et al. Antidepressant-relevant concentrations of the ketamine metabolite (2R,6R)-hydroxynorketamine do not block NMDA receptor function. Proc Nat Acad Sci. 2019;116(11):5160–5169. DOI:10.1073/pnas.1816071116.
  • https://www.auvelity.com/
  • Stahl SM. Dextromethorphan/Bupropion: a novel oral NMDA (N-methyl-D-aspartate) receptor antagonist with multimodal activity. CNS Spectr. 2019;24(5):461–466. Erratum in: CNS Spectr. 25(6), 803 (2020). DOI:10.1017/S1092852919001470.
  • Keam SJ. Dextromethorphan/Bupropion: first approval. CNS Drugs. 2022;36(11):1229–1238.
  • Della Vecchia A, Arone A, Piccinni A, et al. GABA system in depression: impact on pathophysiology and psychopharmacology. Curr Med Chem. 2022;29(36):5710–5730. DOI:10.2174/0929867328666211115124149.
  • Sarawagi A, Soni ND, Patel AB. Glutamate and gaba homeostasis and neurometabolism in major depressive disorder. Front Psychiatry. 2021;12:637863.
  • https://www.zulresso.com/about-zulresso
  • Edinoff AN, Odisho AS, Lewis K, et al. Brexanolone, a GABAA Modulator, in the treatment of postpartum depression in adults: a Comprehensive Review. Front Psychiat. 2021;12:699740.
  • Zhang Q, Dai X, Li W. Comparative efficacy and acceptability of pharmacotherapies for postpartum depression: a systematic review and network meta-analysis. Front Pharmacol. 2022;13:950004.
  • Witkin JM. The mood in the field of antidepressant drug discovery. CNS Neurol Dis Drug Targets. 2011;10(7):1–2.
  • Zhang JC, Yao W, Hashimoto K. Arketamine, a new rapid-acting antidepressant: a historical review and future directions. Neuropharmacology. 2022;218:109219. DOI: 10.1016/j.neuropharm.2022.109219.
  • Leal GC, Bandeira ID, Correia-Melo FS, et al. Intravenous arketamine for treatment-resistant depression: open-label pilot study. Eur Arch Psychiat Clin Neurosci. 2021;271(3):577–582. DOI:10.1007/s00406-020-01110-5.
  • Persson J, Hasselström J, Maurset A, et al. Pharmacokinetics and non-analgesic effects of S- and R-ketamines in healthy volunteers with normal and reduced metabolic capacity. Eur J Clin Pharmacol. 2002;57(12):869–875. DOI:10.1007/s002280100353.
  • Vollenweider FX, Leenders KL, Oye I, et al. Differential psychopathology and patterns of cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using positron emission tomography (PET). Eur Neuropsychopharmacol. 1997;7(1):25–38. DOI:10.1016/S0924-977X(96)00042-9.
  • Malikowska-Racia N, Golebiowska J, Nikiforuk A, et al. Effects of ketamine optical isomers, fluoxetine and naloxone on timing in differential reinforcement of low-rate response (DRL) 72-s task in rats. Eur Neuropsychopharmacol. 2023;67:37–52.
  • Yao W, Cao Q, Luo S, et al. Microglial ERK-NRBP1-CREB-BDNF signaling in sustained antidepressant actions of (R)-ketamine. Mol Psychiatry. 2022;27(3):1618–1629. DOI:10.1038/s41380-021-01377-7
  • Popik P, Khoo SY, Kuziak A, et al. Distinct cognitive and discriminative stimulus effects of ketamine enantiomers in rats. Pharmacol Biochem Behav. 2020;197:173011.
  • Witkin JM, Kranzler J, Kaniecki K, et al. R-(-)-ketamine modifies behavioral effects of morphine predicting efficacy as a novel therapy for opioid use disorder. Pharmacol Biochem Behav. 2020;194:172927.
  • [cited 2023 Feb 10]. Available from: https://ir.atai.life/news-releases/news-release-details/atai-life-sciences-announces-results-phase-2a-trial-pcn-101-r
  • Kato T, Duman RS. Rapastinel, a novel glutamatergic agent with ketamine-like antidepressant actions: convergent mechanisms. Pharmacol Biochem Behavr. 2020;188:172827. doi: 10.1016/j.pbb.2019.172827.
  • Ragguett RM, Tamura JK, McIntyre RS. Keeping up with the clinical advances: depression. CNS Spectr. 2019;24(S1):25–37.
  • https://www.gateneuro.com/
  • Zhang XL, Shuttleworth CW, Moskal JR, et al. Suppression of spreading depolarization and stabilization of dendritic spines by GLYX-13, an NMDA receptor glycine-site functional partial agonist. Exp Neurol. 2015;273:312–321.
  • Highland JN, Zan’s P, Riggs LM, et al. Hydroxynorketamines: pharmacology and potential therapeutic applications. Pharmacol Rev. 2021;73(2):763–791. DOI:10.1124/pharmrev.120.000149. .
  • Bettini E, Stahl SM, De Martin S, 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(8):997. DOI:10.3390/ph15080997
  • Stahl SM, De Martin S, Mattarei A, et al. Esmethadone (REL-1017) and other uncompetitive NMDA channel blockers may improve mood disorders via modulation of synaptic kinase-mediated signaling. Int J Mol Sci. 2022;23(20):12196. DOI:10.3390/ijms232012196.
  • Fava M, Stahl S, Pani L, et al. REL-1017 (Esmethadone) as adjunctive treatment in patients with major depressive disorder: a phase 2a randomized double-blind trial. Amer J Psychiat. 2022;179(2):122–131. DOI:10.1176/appi.ajp.2021.21020197.
  • [cited 2023 Feb 10. Available from: https://www.relmada.com/for-investors/news/detail/272/relmada-therapeutics-announces-top-line-results-from-phase
  • Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacol Biochem Behav. 2022;219:173446.
  • Pałucha-Poniewiera A. The role of mGlu2/3 receptor antagonists in the enhancement of the antidepressant-like effect of ketamine. Pharmacol Biochem Behav. 2022;220:173454.
  • Chaki S, Watanabe M. mGlu2/3 receptor antagonists for depression: overview of underlying mechanisms and clinical development. Eur Arch Psychiat Clin Neurosci. 2023. DOI:10.1007/s00406-023-01561-6. Advance online publication.
  • Witkin JM. mGlu2/3 receptor antagonism: a mechanism to induce rapid antidepressant effects without ketamine-associated side-effects. Pharmacol Biochem Behav. 2020;190:172854.
  • Watanabe M, Marcy B, Hiroki A, 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. Int J Neuropsychopharmacol. 2022;25(2):106–117. DOI:10.1093/ijnp/pyab062.
  • Alt A, Nisenbaum ES, Bleakman D, et al. A role for AMPA receptors in mood disorders. Biochem Pharmacol. 2006;71:1273–1288. .
  • Kadriu B, Musazzi L, Johnston JN, et al. Positive AMPA receptor modulation in the treatment of neuropsychiatric disorders: a long and winding road. Drug Discov Today. 2021;26(12):2816–2838. DOI:10.1016/j.drudis.2021.07.027
  • Nations KR, Dogterom P, Bursi R, et al. Examination of Org 26576, an AMPA receptor positive allosteric modulator, in patients diagnosed with major depressive disorder: an exploratory, randomized, double-blind, placebo-controlled trial. J Psychopharmacol. 2012;26:1525–1539.
  • Hara H, Suzuki A, Kunugi A, et al. TAK-653, an AMPA receptor potentiator with minimal agonistic activity, produces an antidepressant-like effect with a favorable safety profile in rats. Pharmacol Biochem Behav. 2021;211:173289.
  • http://respirerx.com/product-pipeline/
  • Kato AS, Witkin JM. Auxiliary subunits of AMPA receptors: the discovery of a forebrain-selective antagonist, LY3130481/CERC-611. Biochem Pharmacol. 2018;147:191–200.
  • Herguedas B, Kohegyi BK, Dohrke JN, et al. Mechanisms underlying TARP modulation of the GluA1/2-γ8 AMPA receptor. Nat Commun. 2022;13(1):734. DOI:10.1038/s41467-022-28404-7.
  • Zhang D, Lape R, Shaikh SA, et al. Modulatory mechanisms of TARP γ8-selective AMPA receptor therapeutics. Nat Commun. 2023;14:1659.
  • Witkin JM, Smith JL, Golani LK. Involvement of muscarinic receptor mechanisms in antidepressant drug action. Adv Pharmacol. 2020;89:311–356.
  • Griffiths RR, Richards WA, McCann U, et al. Psilocybin can occasion mystical-type experiences having substantial and sustained personal meaning and spiritual significance. Psychopharmacology. 2006;187(3):268–292. DOI:10.1007/s00213-006-0457-5.
  • Studerus E, Kometer M, Hasler F, et al. Acute, subacute and long term subjective effects of psilocybin in healthy humans: a pooled analysis of experimental studies. J Psychopharmacol. 2011;25:1434–1452.
  • Grob CS, Danforth AL, Chopra GS, et al. Pilot study of psilocybin treatment for anxiety in patients with advanced stage cancer. Archs Gen Psychiat. 2011;68(1):71–78. DOI:10.1001/archgenpsychiatry.2010.116.
  • Davis AK, Barrett FS, May DG, et al. Effects of psilocybin-assisted therapy on major depressive disorder: a randomized clinical trial. JAMA Psychiatry. 2021;78(5):481–489. Erratum in: JAMA Psychiatry, PMID: 33146667 (2021). DOI:10.1001/jamapsychiatry.2020.3285.
  • Goel DB, Zilate S. Potential therapeutic effects of psilocybin: a systematic review. Cureus. 2022;14(10):e30214. DOI:10.7759/cureus.30214.
  • Ledwos N, Rosenblat JD, Blumberger DM, et al. A critical appraisal of evidence on the efficacy and safety of serotonergic psychedelic drugs as emerging antidepressants: mind the evidence gap. J Clin Psychopharm. 2022;42(6):581–588. DOI:10.1097/JCP.0000000000001608
  • White CM, Weisman N, Dalo J. Psychedelics for patients with cancer: a comprehensive literature review. Ann Pharmacother. 2023;10600280221144055. DOI:10.1177/10600280221144055
  • Zeiss R, Gahr M, Graf H. Rediscovering psilocybin as an antidepressive treatment strategy. Pharmaceuticals. 2021;14(10):985.
  • https://www.gilgameshpharmaceutical.com/pipeline
  • Mathai DS, Nayak SM, Yaden DB, et al. Reconsidering “dissociation” as a predictor of antidepressant efficacy for esketamine. Psychopharmacology. 2023;240:827–836.
  • Murray CH, Tare I, Perry CM, et al. Low doses of LSD reduce broadband oscillatory power and modulate event-related potentials in healthy adults. Psychopharmacology. 2022;239(6):1735–1747. DOI:10.1007/s00213-021-05991-9
  • Qu Y, Chang L, Ma L, et al. Rapid antidepressant-like effect of non-hallucinogenic psychedelic analog lisuride, but not hallucinogenic psychedelic DOI, in lipopolysaccharide-treated mice. Pharmacol Biochem Behav. 2023;222:173500.
  • [cited 2023 Mar 5]. Available from: https://www.healio.com/news/psychiatry/20230208/fda-accepts-application-for-zuranolone-to-treat-depression#:~:text=The%20FDA%20has%20accepted%20a,a%20release%20from%20the%20manufacturers
  • Fee C, Prevot TD, Misquitta K, et al. Behavioral deficits induced by somatostatin-positive gaba neuron silencing are rescued by alpha 5 gaba-a receptor potentiation. Int J Neuropsychopharmacol. 2021;24(6):505–518. DOI:10.1093/ijnp/pyab002.
  • Troppoli TA, Zanos P, Georgiou P, et al. Negative allosteric modulation of gamma-aminobutyric acid a receptors at α5 subunit-containing benzodiazepine sites reverses stress-induced anhedonia and weakened synaptic function in mice. Biol Psychiatry. 2022;92(3):216–226. DOI:10.1016/j.biopsych.2021.11.024.
  • Methuku KR, Li X, Cerne R, et al. An antidepressant-related pharmacological signature for positive allosteric modulators of α2/3-containing GABAA receptors. Pharmacol Biochem Behav. 2018;170:9–13.
  • Witkin JM, Cerne R, Wakulchik M, et al. Further evaluation of the potential anxiolytic activity of imidazo[1,5-a][1,4]diazepin agents selective for α2/3-containing GABAA receptors. Pharmacol Biochem Behav. 2017;157:35–40.
  • Branchi I, Poggini S, Capuron L, et al. Brain-immune crosstalk in the treatment of major depressive disorder. Eur Neuropsychopharmacol. 2021;45:89–107.
  • Ruiz NAL, Del Ángel DS, Brizuela NO, et al. Inflammatory process and immune system in major depressive disorder. Int J Neuropsychopharmacol. 2022;25(1):46–53. DOI:10.1093/ijnp/pyab072.
  • Sørensen NV, Frandsen BH, Orlovska-Waast S, et al. Immune cell composition in unipolar depression: a comprehensive systematic review and meta-analysis. Mol Psychiatry. 2023;28(1):391–401. DOI:10.1038/s41380-022-01905-z.
  • Tecoma ES, Huey LY. Psychic distress and the immune response. Life Sci. 1985;36(19):1799–1812.
  • Duarte-Silva E, de Melo MG, Maes M, et al. Shared metabolic and neuroimmune mechanisms underlying Type 2 diabetes mellitus and major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2021;111:110351.
  • Witkin JM. The romantic age of pharmacological science. Pharmacol Biochem Behav. 2022;214:173354.
  • Domino EF, Warner DS. Taming the ketamine tiger. Anesthesiology. 2010;113(3):678–684.
  • Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial. Arch Gen Psychiatry. 2006;63(10):1121–1129. DOI:10.1001/archpsyc.63.10.1121.
  • Lapidos A, Lopez-Vives D, Sera CE, et al. Patients’ recovery and non-recovery narratives after intravenous ketamine for treatment-resistant depression. J Affective Dis. 2023;323:534–539.
  • Bershad AK, Preller KH, Lee R, et al. Preliminary report on the effects of a low dose of LSD on resting-state amygdala functional connectivity. Biol Psychiat: Cogn Neurosci Neuroimag. 2020;5(4):461–467. DOI:10.1016/j.bpsc.2019.12.007
  • Wong A, Raz A. Microdosing with classical psychedelics: research trajectories and practical considerations. Transcultural Psychiat. 2022;59(5):675–690.
  • Cavanna F, Muller S, de la Fuente LA, et al. Microdosing with psilocybin mushrooms: a double-blind placebo-controlled study. Trans Psychiat. 2022;12(1):307. DOI:10.1038/s41398-022-02039-0
  • Rootman JM, Kiraga M, Kryskow P, et al. Psilocybin microdosers demonstrate greater observed improvements in mood and mental health at one month relative to non-microdosing controls. Sci Rep. 2022;12(1):11091.
  • Polito V, Liknaitzky P. The emerging science of microdosing: a systematic review of research on low dose psychedelics (1955-2021) and recommendations for the field. Neurosci Biobehav Rev. 2022;139:104706.
  • Roy B, Dwivedi Y. An insight into the sprawling microverse of microRnas in depression pathophysiology and treatment response. Neurosci Biobehav Rev. 2023;146:105040.
  • Cohen SE, Zantvoord JB, Wezenberg BN, et al. Electroencephalography for predicting antidepressant treatment success: a systematic review and meta-analysis. J Affec Dis. 2023;321:201–207.
  • Watts D, Pulice RF, Reilly J, et al. Predicting treatment response using EEG in major depressive disorder: a machine-learning meta-analysis. Transl Psychiatry. 2022;12(1):332. DOI:10.1038/s41398-022-02064-z
  • Gerlach AR, Karim HT, Peciña M, et al. MRI predictors of pharmacotherapy response in major depressive disorder. NeuroImage Clin. 2022;36:103157.
  • Micale V, Di Bartolomeo M, Di Martino S, et al. Are the epigenetic changes predictive of therapeutic efficacy for psychiatric disorders? A translational approach towards novel drug targets. Pharmacol Therap. 2023;241:108279.
  • Gasparini A, Callegari C, Lucca G, et al. Inflammatory biomarker and response to antidepressant in major depressive disorder: a systematic review and meta-analysis. Psychopharm Bull. 2022;52(1):36–52.
  • Foster JA, Baker GB, Dursun SM. The relationship between the gut microbiome-immune system-brain axis and major depressive disorder. Front Neurol. 2021;12:721126.
  • Jambor T, Juhasz G, Eszlari N. Towards personalised antidepressive medicine based on “big data”: an up-to-date review on robust factors affecting treatment response. Neuropsychopharmacologia Hungarica. 2022;24(1):17–28.
  • Alario AA, Niciu MJ. Biomarkers of ketamine’s antidepressant effect: a clinical review of genetics, functional connectivity, and neurophysiology. Chronic Stress. 2021;5:24705470211014210.
  • Meshkat S, Ho RC, Cao B, et al. Biomarkers of ketamine’s antidepressant effect: an umbrella review. J Affect Dis. 2023;323:598–606.
  • Jiang Y, Zhang T, Zhang M, et al. Apathy in melancholic depression and abnormal neural activity within the reward-related circuit. Behav Brain Res. 2023;444:114379.
  • Li X, Zhang H, Han X, et al. Predictive potential of somatic symptoms for the identification of subthreshold depression and major depressive disorder in primary care settings. meta-analysis. J Affect Disord. 2023;205:1–12.
  • Rajewska-Rager A, Dmitrzak-Weglarz M, Lepczynska N, et al. Dimensions of the Hamilton Depression Rating Scale correlate with impulsivity and personality traits among youth patients with depression. J Clin Med. 2023;12(5):1744. DOI:10.3390/jcm12051744.
  • Suseelan S, Pinna G. Heterogeneity in major depressive disorder: the need for biomarker-based personalized treatments. Adv Clin Chem. 2023;112:1–67.
  • Kropp DR, Hodes GE. Sex differences in depression: an immunological perspective. Brain Res Bull. 2023;196:34–45.
  • Strawn JR, Mills JA, Suresh V, et al. The impact of age on antidepressant response: a mega-analysis of individuals with major depressive disorder. J Psychiatr Res. 2023;159:266–273.
  • Fornaro M, Aguglia E, Dell’osso L, et al. Could the underestimation of bipolarity obstruct the search for novel antidepressant drugs? Expert Opin Pharmacother. 2011;12(18):2817–2831. DOI:10.1517/14656566.2011.632366
  • Stein K, Maruf AA, Müller DJ, et al. Serotonin transporter genetic variation and antidepressant response and tolerability: a systematic review and meta-analysis. J Pers Med. 2021;11(12):1334. DOI:10.3390/jpm11121334.
  • Jakobsen JC, Katakam KK, Schou A, et al. Selective serotonin reuptake inhibitors versus placebo in patients with major depressive disorder. A systematic review with meta-analysis and Trial Sequential Analysis. BMC Psychiatry. 2017;17(1):58. DOI:10.1186/s12888-016-1173-2.
  • Tham A, Jonsson U, Andersson G, et al. Efficacy and tolerability of antidepressants in people aged 65 years or older with major depressive disorder - a systematic review and a meta-analysis. J Affect Disord. 2016;205:1–12.
  • Harkness KL, Chakrabarty T, Rizvi SJ, et al. the differential relation of emotional, physical, and sexual abuse histories to antidepressant treatment remission and persistence of anhedonia in major depression: a can-bind-1 report. Can J Psychiatry. Feb 2023;13:7067437231156255. Epub ahead of print. PMID: 36785892. DOI:10.1177/07067437231156255.
  • Bespalov A, Steckler T, Altevogt B, et al. Failed trials for central nervous system disorders do not necessarily invalidate preclinical models and drug targets. Nat Rev Drug Discov. 2016;15(7):516. DOI:10.1038/nrd.2016.88
  • Rodrigues M, Syed Z, Dufort A, et al. Heterogeneity across outcomes reported in clinical trials for older adults with depression: a systematic survey. J Clin Epidemiol. 2023;S0895-4356(23):00045–8.
  • Křenek P, Hořínková J, Bartečků E. Peripheral inflammatory markers in subtypes and core features of depression: a systematized review. Psychopathology. 2023;22:1–14.
  • Calder AE, Hasler G. Towards an understanding of psychedelic-induced neuroplasticity. Neuropsychopharmacology. 2023;48(1):104–112.

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.