Skip to content
2000
image of Exploring New Frontiers in Pharmacological Treatment of Depression: A Review on Recent Advances

Abstract

The current treatment strategy is still subpar, especially for severe mental problems, despite tremendous progress in the understanding of the central nervous system. Improving healthcare usually entails two main approaches: investigating new treatment approaches and improving current ones. New pharmacological options include enhanced monoaminergic pharmaceuticals, old treatments reassessed with a better knowledge of the biology of mental disease, and medications that target new therapeutic pathways. One major clinical challenge in the treatment of depression is resistance to antidepressant drugs. It appears promising to switch to monotherapy using new multifunctional antidepressants and add new atypical antipsychotics, such as brexpiprazole and aripiprazole. Current efforts are concentrated on unraveling depression's origins and pinpointing fresh targets for pharmacological intervention. This review explores encouraging novel pharmacological avenues for major depressive disorder treatment. These include targeting receptors, such as N-methyl-D-aspartate and metabotropic glutamate receptors, and employing glutamatergic modulators and various augmentation strategies, all of which hold the potential for reversal of depressant effects. Combining innovative concepts with enhancements of existing discoveries may propel antidepressant research forward, offering hope for developing compounds that are effective and rapid in their action, even among patients who have found limited success with other therapies.

Loading

Article metrics loading...

/content/journals/cmc/10.2174/0109298673342524250109181220
2025-05-22
2025-11-06
Loading full text...

Full text loading...

References

  1. Smith K. Torres D.I. Mental health: A world of depression. Nature 2014 515 7526 180 181 10.1038/515180a 25391942
    [Google Scholar]
  2. Mathers C.D. Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006 3 11 e442 10.1371/journal.pmed.0030442 17132052
    [Google Scholar]
  3. Chisholm D. Sweeny K. Sheehan P. Rasmussen B. Smit F. Cuijpers P. Saxena S. Scaling-up treatment of depression and anxiety: A global return on investment analysis. Lancet Psychiatry 2016 3 5 415 424 10.1016/S2215‑0366(16)30024‑4 27083119
    [Google Scholar]
  4. Hanssen H. Minghetti A. Faude O. Trucksäss S.A. Zahner L. Beck J. Donath L. Effects of endurance exercise modalities on arterial stiffness in patients suffering from unipolar depression: A randomized controlled trial. Front. Psychiatry 2018 8 311 10.3389/fpsyt.2017.00311 29403399
    [Google Scholar]
  5. Kvam S. Kleppe C.L. Nordhus I.H. Hovland A. Exercise as a treatment for depression: A meta-analysis. J. Affect. Disord. 2016 202 67 86 10.1016/j.jad.2016.03.063 27253219
    [Google Scholar]
  6. Delgado P.L. Depression: The case for a monoamine deficiency. J. Clin. Psychiatry 2000 61 6 Suppl. 6 7 11 10775018
    [Google Scholar]
  7. Edinoff A.N. Akuly H.A. Hanna T.A. Ochoa C.O. Patti S.J. Ghaffar Y.A. Kaye A.D. Viswanath O. Urits I. Boyer A.G. Cornett E.M. Kaye A.M. Selective serotonin reuptake inhibitors and adverse effects: A narrative review. Neurol. Int. 2021 13 3 387 401 10.3390/neurolint13030038 34449705
    [Google Scholar]
  8. Vieira M.R. Salvadore G. Luckenbaugh D.A. Manji H.K. Zarate C.A. Jr Rapid onset of antidepressant action: A new paradigm in the research and treatment of major depressive disorder. J. Clin. Psychiatry 2008 69 6 946 958 10.4088/JCP.v69n0610 18435563
    [Google Scholar]
  9. Kamran M. Bibi F. Rehman U.A. Morris D.W. ur. Rehman A, Morris DW. Major depressive disorder: Existing hypotheses about pathophysiological mechanisms and new genetic findings. Genes (Basel) 2022 13 4 646 10.3390/genes13040646 35456452
    [Google Scholar]
  10. Koenigsberg JZ Depressive disorders: Integrated and unified psychotherapy approaches. Taylor & Francis Milton Park, Abingdon in the United Kingdom 2023
    [Google Scholar]
  11. Cardon I. Grobecker S. Kücükoktay S. Bader S. Jahner T. Nothdurfter C. Koschitzki K. Berneburg M. Weber B.H.F. Stöhr H. Höring M. Liebisch G. Braun F. Hampl R.T. Riemenschneider M.J. Rupprecht R. Milenkovic V.M. Wetzel C.H. Mitochondrial and cellular function in fibroblasts, induced neurons, and astrocytes derived from case study patients: Insights into major depression as a mitochondria-associated disease. Int. J. Mol. Sci. 2024 25 2 963 10.3390/ijms25020963 38256041
    [Google Scholar]
  12. Artigas F. Developments in the field of antidepressants, where do we go now? Eur. Neuropsychopharmacol. 2015 25 5 657 670 10.1016/j.euroneuro.2013.04.013 23706576
    [Google Scholar]
  13. Gadad B.S. Jha M.K. Czysz A. Furman J.L. Mayes T.L. Emslie M.P. Trivedi M.H. Peripheral biomarkers of major depression and antidepressant treatment response: Current knowledge and future outlooks. J. Affect. Disord. 2018 233 3 14 10.1016/j.jad.2017.07.001 28709695
    [Google Scholar]
  14. Rakesh G. Pae C.U. Masand P.S. Beyond serotonin: Newer antidepressants in the future. Expert Rev. Neurother. 2017 17 8 777 790 10.1080/14737175.2017.1341310 28598698
    [Google Scholar]
  15. Pilc A. Chaki S. Nowak G. Witkin J.M. Mood disorders: Regulation by metabotropic glutamate receptors. Biochem. Pharmacol. 2008 75 5 997 1006 10.1016/j.bcp.2007.09.021 18164691
    [Google Scholar]
  16. Dogra S. Conn P.J. Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 2021 196 108687 10.1016/j.neuropharm.2021.108687 34175327
    [Google Scholar]
  17. Chaki S. Koike H. Fukumoto K. Targeting of metabotropic glutamate receptors for the development of novel antidepressants. Chronic. Stress. 2019 3 2470547019837712 10.1177/2470547019837712 32500107
    [Google Scholar]
  18. Yüksel C. Öngür D. Magnetic resonance spectroscopy studies of glutamate-related abnormalities in mood disorders. Biol. Psychiatry 2010 68 9 785 794 10.1016/j.biopsych.2010.06.016 20728076
    [Google Scholar]
  19. Li C.T. Yang K.C. Lin W.C. Glutamatergic dysfunction and glutamatergic compounds for major psychiatric disorders: Evidence from clinical neuroimaging studies. Front. Psychiatry 2019 9 767 10.3389/fpsyt.2018.00767 30733690
    [Google Scholar]
  20. Poletti S. Mazza M.G. Vai B. Lorenzi C. Colombo C. Benedetti F. Proinflammatory cytokines predict brain metabolite concentrations in the anterior cingulate cortex of patients with bipolar disorder. Front. Psychiatry 2020 11 590095 10.3389/fpsyt.2020.590095 33363485
    [Google Scholar]
  21. Lodge D. Mercier M.S. Ketamine and phencyclidine: The good, the bad and the unexpected. Br. J. Pharmacol. 2015 172 17 4254 4276 10.1111/bph.13222 26075331
    [Google Scholar]
  22. Serafini G. Howland R. Rovedi F. Girardi P. Amore M. The role of ketamine in treatment-resistant depression: A systematic review. Curr. Neuropharmacol. 2014 12 5 444 461 10.2174/1570159X12666140619204251 25426012
    [Google Scholar]
  23. Yamashita F. Hashida M. Pharmacokinetic considerations for targeted drug delivery. Adv. Drug Deliv. Rev. 2013 65 1 139 147 10.1016/j.addr.2012.11.006 23280371
    [Google Scholar]
  24. Borbély É. Simon M. Fuchs E. Wiborg O. Czéh B. Helyes Z. Novel drug developmental strategies for treatment-resistant depression. Br. J. Pharmacol. 2022 179 6 1146 1186 10.1111/bph.15753 34822719
    [Google Scholar]
  25. Bibi Z. Role of cytochrome P450 in drug interactions. Nutr. Metab. 2008 5 1 27 10.1186/1743‑7075‑5‑27 18928560
    [Google Scholar]
  26. Wang J. Goffer Y. Xu D. Tukey D.S. Shamir D.B. Eberle S.E. Zou A.H. Blanck T.J.J. Ziff E.B. A single subanesthetic dose of ketamine relieves depression-like behaviors induced by neuropathic pain in rats. Anesthesiology 2011 115 4 812 821 10.1097/ALN.0b013e31822f16ae 21934410
    [Google Scholar]
  27. Kohtala S. Ketamine—50 years in use: From anesthesia to rapid antidepressant effects and neurobiological mechanisms. Pharmacol. Rep. 2021 73 2 323 345 10.1007/s43440‑021‑00232‑4 33609274
    [Google Scholar]
  28. Krystal J.H. Kavalali E.T. Monteggia L.M. Ketamine and rapid antidepressant action: New treatments and novel synaptic signaling mechanisms. Neuropsychopharmacology 2024 49 1 41 50 10.1038/s41386‑023‑01629‑w 37488280
    [Google Scholar]
  29. An X. Yao X. Li B. Yang W. Cui R. Zhao G. Jin Y. Role of BDNF-mTORC1 signaling pathway in female depression. Neural Plast. 2021 2021 1 8 10.1155/2021/6619515 33628219
    [Google Scholar]
  30. Peyrovian B. Rosenblat J.D. Pan Z. Iacobucci M. Brietzke E. McIntyre R.S. The glycine site of NMDA receptors: A target for cognitive enhancement in psychiatric disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019 92 387 404 10.1016/j.pnpbp.2019.02.001 30738126
    [Google Scholar]
  31. Henter I.D. Sousa d.R.T. Zarate C.A. Jr Glutamatergic modulators in depression. Harv. Rev. Psychiatry 2018 26 6 307 319 10.1097/HRP.0000000000000183 29465478
    [Google Scholar]
  32. Levy H.U. Javitt D.C. Gelfin Y. Gorelik E. Bar M. Blanaru M. Kremer I. Controlled trial of d-cycloserine adjuvant therapy for treatment-resistant major depressive disorder. J. Affect. Disord. 2006 93 1-3 239 243 10.1016/j.jad.2006.03.004 16677714
    [Google Scholar]
  33. Lee E.E. Selva D.M.P. Liu A. Himelhoch S. Ketamine as a novel treatment for major depressive disorder and bipolar depression: A systematic review and quantitative meta-analysis. Gen. Hosp. Psychiatry 2015 37 2 178 184 10.1016/j.genhosppsych.2015.01.003 25698228
    [Google Scholar]
  34. Henter I.D. Park L.T. Zarate C.A. Jr Novel glutamatergic modulators for the treatment of mood disorders: Current status. CNS Drugs 2021 35 5 527 543 10.1007/s40263‑021‑00816‑x 33904154
    [Google Scholar]
  35. Vecera C.M. Courtes C.A.
  36. Asghar J. Tabasam M. Althobaiti M.M. Ashour A.A. Aleid M.A. Khalaf I.O. Aldhyani T.H.H. A randomized clinical trial comparing two treatment strategies, evaluating the meaningfulness of HAM-D rating scale in patients with major depressive disorder. Front. Psychiatry 2022 13 873693 10.3389/fpsyt.2022.873693 35722557
    [Google Scholar]
  37. Fenton C. McLoughlin D.M. Usefulness of hamilton rating scale for depression subset scales and full versions for electroconvulsive therapy. PLoS One 2021 16 11 e0259861 10.1371/journal.pone.0259861 34752484
    [Google Scholar]
  38. MahmoudianDehkordi S. Ahmed A.T. Bhattacharyya S. Han X. Baillie R.A. Arnold M. Skime M.K. Williams J.L.S. Moseley M.A. Thompson J.W. Louie G. Posse R.P. Craighead W.E. McDonald W. Krishnan R. Rush A.J. Frye M.A. Dunlop B.W. Weinshilboum R.M. Daouk K.R. Daouk K.R. Rush J. Tenenbaum J. Moseley A. Thompson W. Louie G. Blach C. Mahmoudiandehkhordi S. Baillie R. Han X. Bhattacharyya S. Frye M. Weinshilboum R. Ahmed A. Neavin D. Liu D. Skime M. Rinaldo P. Fiehn O. Brydges C. Mayberg H. Choi K.S. Cha J. Kastenmüller G. Arnold M. Binder E. Arloth K.J. Holgado N.A. Shi L. Dunlop B. Craighead E. McDonald W. Posse P.R. Penninx B. Milaneschi Y. Jansen R. Krishnan R. Alterations in acylcarnitines, amines, and lipids inform about the mechanism of action of citalopram/escitalopram in major depression. Transl. Psychiatry. 2021 11 1 153 10.1038/s41398‑020‑01097‑6 33654056
    [Google Scholar]
  39. Bourque M. Grégoire L. Patel W. Dickens D. Snodgrass R. Paolo D.T. AV-101, a pro-drug antagonist at the nmda receptor glycine site, reduces l-dopa induced dyskinesias in mptp monkeys. Cells 2022 11 22 3530 10.3390/cells11223530 36428960
    [Google Scholar]
  40. Park L.T. Kadriu B. Gould T.D. Zanos P. Greenstein D. Evans J.W. Yuan P. Farmer C.A. Oppenheimer M. George J.M. Adeojo L.W. Snodgrass H.R. Smith M.A. Henter I.D. Vieira M.R. Mannes A.J. Zarate C.A. Jr A randomized trial of the n-methyl-d-aspartate receptor glycine site antagonist prodrug 4-chlorokynurenine in treatment-resistant depression. Int. J. Neuropsychopharmacol. 2020 23 7 417 425 10.1093/ijnp/pyaa025 32236521
    [Google Scholar]
  41. Hecking J. Davoudian P.A. Wilkinson S.T. Emerging therapeutics based on the amino acid neurotransmitter system: An update on the pharmaceutical pipeline for mood disorders. Chronic. Stress. 2021 5 24705470211020446 10.1177/24705470211020446 34124495
    [Google Scholar]
  42. Willard S.S. Koochekpour S. Glutamate, glutamate receptors, and downstream signaling pathways. Int. J. Biol. Sci. 2013 9 9 948 959 10.7150/ijbs.6426 24155668
    [Google Scholar]
  43. Schmidt H.D. Pierce R.C. Cocaine-induced neuroadaptations in glutamate transmission. Ann. N. Y. Acad. Sci. 2010 1187 1 35 75 10.1111/j.1749‑6632.2009.05144.x 20201846
    [Google Scholar]
  44. Wolf M.E. Ferrario C.R. AMPA receptor plasticity in the nucleus accumbens after repeated exposure to cocaine. Neurosci. Biobehav. Rev. 2010 35 2 185 211 10.1016/j.neubiorev.2010.01.013 20109488
    [Google Scholar]
  45. Jaso B.A. Niciu M.J. Iadarola N.D. Lally N. Richards E.M. Park M. Ballard E.D. Nugent A.C. Vieira M.R. Zarate C.A. Therapeutic modulation of glutamate receptors in major depressive disorder. Curr. Neuropharmacol. 2017 15 1 57 70 10.2174/1570159X14666160321123221 26997505
    [Google Scholar]
  46. Henter I.D. Sousa d.R.T. Gold P.W. Brunoni A.R. Zarate C.A. Jr Vieira M.R. Mood therapeutics: Novel pharmacological approaches for treating depression. Expert Rev. Clin. Pharmacol. 2017 10 2 153 166 10.1080/17512433.2017.1253472 27781556
    [Google Scholar]
  47. Riveros C. Dechartres A. Perrodeau E. Haneef R. Boutron I. Ravaud P. Timing and completeness of trial results posted at ClinicalTrials.gov and published in journals. PLoS Med. 2013 10 12 e1001566 10.1371/journal.pmed.1001566 24311990
    [Google Scholar]
  48. Dechartres A. Bond E.G. Scheer J. Riveros C. Atal I. Ravaud P. Reporting of statistically significant results at ClinicalTrials.gov for completed superiority randomized controlled trials. BMC Med. 2016 14 1 192 10.1186/s12916‑016‑0740‑1 27899150
    [Google Scholar]
  49. Kent J.M. Daly E. Kezic I. Lane R. Lim P. Smedt D.H. Boer D.P. Nueten V.L. Drevets W.C. Ceusters M. Efficacy and safety of an adjunctive mGlu2 receptor positive allosteric modulator to a SSRI/SNRI in anxious depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2016 67 66 73 10.1016/j.pnpbp.2016.01.009 26804646
    [Google Scholar]
  50. Lavreysen H. Ahnaou A. Drinkenburg W. Langlois X. Mackie C. Pype S. Lütjens R. Poul L.E. Trabanco A.A. Nuñez J.M.C. Pharmacological and pharmacokinetic properties of JNJ-40411813, a positive allosteric modulator of the mGlu2 receptor. Pharmacol. Res. Perspect. 2015 3 1 e00096 10.1002/prp2.96 25692015
    [Google Scholar]
  51. Rose M. Devine J. Assessment of patient-reported symptoms of anxiety. Dialogues Clin. Neurosci. 2014 16 2 197 211 10.31887/DCNS.2014.16.2/mrose 25152658
    [Google Scholar]
  52. Kocarnik J.M. Compton K. Dean F.E. Fu W. Gaw B.L. Harvey J.D. Henrikson H.J. Lu D. Pennini A. Xu R. Ababneh E. Kangevari A.M. Abbastabar H. Elsalam A.S.M. Abdoli A. Abedi A. Abidi H. Abolhassani H. Adedeji I.A. Adnani Q.E.S. Advani S.M. Afzal M.S. Aghaali M. Ahinkorah B.O. Ahmad S. Ahmad T. Ahmadi A. Ahmadi S. Rashid A.T. Salih A.Y. Akalu G.T. Aklilu A. Akram T. Akunna C.J. Hamad A.H. Alahdab F. Aly A.Z. Ali S. Alimohamadi Y. Alipour V. Aljunid S.M. Alkhayyat M. Hashiani A.A. Almasri N.A. Maweri A.S.A.A. Almustanyir S. Alonso N. Guzman A.N. Amu H. Anbesu E.W. Ancuceanu R. Ansari F. Moghaddam A.A. Antwi M.H. Anvari D. Anyasodor A.E. Aqeel M. Arabloo J. Zozani A.M. Aremu O. Ariffin H. Aripov T. Arshad M. Artaman A. Arulappan J. Asemi Z. Jafarabadi A.M. Ashraf T. Atorkey P. Aujayeb A. Ausloos M. Awedew A.F. Quintanilla A.B.P. Ayenew T. Azab M.A. Azadnajafabad S. Jafari A.A. Azarian G. Azzam A.Y. Badiye A.D. Bahadory S. Baig A.A. Baker J.L. Balakrishnan S. Banach M. Bärnighausen T.W. Adesi B.F. Barra F. Barrow A. Behzadifar M. Belgaumi U.I. Bezabhe W.M.M. Bezabih Y.M. Bhagat D.S. Bhagavathula A.S. Bhardwaj N. Bhardwaj P. Bhaskar S. Bhattacharyya K. Bhojaraja V.S. Bibi S. Bijani A. Biondi A. Bisignano C. Bjørge T. Bleyer A. Blyuss O. Bolarinwa O.A. Bolla S.R. Braithwaite D. Brar A. Brenner H. Teixeira B.M.T. Butt N.S. Butt Z.A. Caetano dos Santos F.L. Cao Y. Carreras G. López C.F. Cembranel F. Cerin E. Cernigliaro A. Chakinala R.C. Chattu S.K. Chattu V.K. Chaturvedi P. Ochir C.O. Cho D.Y. Christopher D.J. Chu D.T. Chung M.T. Conde J. Cortés S. Cortesi P.A. Costa V.M. Cunha A.R. Dadras O. Dagnew A.B. Dahlawi S.M.A. Dai X. Dandona L. Dandona R. Darwesh A.M. Neves d.J. De la Hoz F.P. Demis A.B. Gutiérrez D.E. Dhamnetiya D. Dhimal M.L. Dhimal M. Dianatinasab M. Diaz D. Djalalinia S. Do H.P. Doaei S. Dorostkar F. dos Santos Figueiredo F.W. Driscoll T.R. Ebrahimi H. Eftekharzadeh S. Tantawi E.M. Abid E.H. Elbarazi I. Elhabashy H.R. Elhadi M. Jaafary E.S.I. Eshrati B. Eskandarieh S. Esmaeilzadeh F. Etemadi A. Ezzikouri S. Faisaluddin M. Faraon E.J.A. Fares J. Farzadfar F. Feroze A.H. Ferrero S. Desideri F.L. Filip I. Fischer F. Fisher J.L. Foroutan M. Fukumoto T. Gaal P.A. Gad M.M. Gadanya M.A. Gallus S. Fonseca G.M. Obsa G.A. Ghafourifard M. Ghashghaee A. Ghith N. Gholamalizadeh M. Gilani S.A. Ginindza T.G. Gizaw A.T.T. Glasbey J.C. Golechha M. Goleij P. Gomez R.S. Gopalani S.V. Gorini G. Goudarzi H. Grosso G. Gubari M.I.M. Guerra M.R. Guha A. Gunasekera D.S. Gupta B. Gupta V.B. Gupta V.K. Gutiérrez R.A. Nejad H.N. Haider M.R. Mirzaian H.A. Halwani R. Hamadeh R.R. Hameed S. Hamidi S. Hanif A. Haque S. Harlianto N.I. Haro J.M. Hasaballah A.I. Hassanipour S. Hay R.J. Hay S.I. Hayat K. Heidari G. Heidari M. Serna H.B.Y. Herteliu C. Hezam K. Holla R. Hossain M.M. Hossain M.B.H. Hosseini M.S. Hosseini M. Hosseinzadeh M. Hostiuc M. Hostiuc S. Househ M. Hsairi M. Huang J. Hugo F.N. Hussain R. Hussein N.R. Hwang B.F. Iavicoli I. Ibitoye S.E. Ida F. Ikuta K.S. Ilesanmi O.S. Ilic I.M. Ilic M.D. Irham L.M. Islam J.Y. Islam R.M. Islam S.M.S. Ismail N.E. Isola G. Iwagami M. Jacob L. Jain V. Jakovljevic M.B. Javaheri T. Jayaram S. Jazayeri S.B. Jha R.P. Jonas J.B. Joo T. Joseph N. Joukar F. Jürisson M. Kabir A. Kahrizi D. Kalankesh L.R. Kalhor R. Kaliyadan F. Kalkonde Y. Kamath A. Kameran Al-Salihi N. Kandel H. Kapoor N. Karch A. Kasa A.S. Katikireddi S.V. Kauppila J.H. Kavetskyy T. Kebede S.A. Keshavarz P. Keykhaei M. Khader Y.S. Khalilov R. Khan G. Khan M. Khan M.N. Khan M.A.B. Khang Y.H. Khater A.M. Khayamzadeh M. Kim G.R. Kim Y.J. Kisa A. Kisa S. Skarbek K.K. Kopec J.A. Koteeswaran R. Koul P.A. Laxminarayana K.S.L. Koyanagi A. Bicer K.B. Kugbey N. Kumar G.A. Kumar N. Kumar N. Kurmi O.P. Kutluk T. Vecchia L.C. Lami F.H. Landires I. Lauriola P. Lee S. Lee S.W.H. Lee W.C. Lee Y.H. Leigh J. Leong E. Li J. Li M.C. Liu X. Loureiro J.A. Lunevicius R. Magdy Abd El Razek M. Majeed A. Makki A. Male S. Malik A.A. Mansournia M.A. Martini S. Masoumi S.Z. Mathur P. McKee M. Mehrotra R. Mendoza W. Menezes R.G. Mengesha E.W. Mesregah M.K. Mestrovic T. Jonasson M.J. Miazgowski B. Miazgowski T. Michalek I.M. Miller T.R. Mirzaei H. Mirzaei H.R. Misra S. Mithra P. Moghadaszadeh M. Mohammad K.A. Mohammad Y. Mohammadi M. Mohammadi S.M. Hafshejani M.A. Mohammed S. Moka N. Mokdad A.H. Molokhia M. Monasta L. Moni M.A. Moosavi M.A. Moradi Y. Moraga P. Morgado-da-Costa J. Morrison S.D. Mosapour A. Mubarik S. Mwanri L. Nagarajan A.J. Nagaraju S.P. Nagata C. Naimzada M.D. Nangia V. Naqvi A.A. Swamy N.S. Ndejjo R. Nduaguba S.O. Negoi I. Negru S.M. Kandel N.S. Nguyen C.T. Nguyen H.L.T. Niazi R.K. Nnaji C.A. Noor N.M. Samudio N.V. Nzoputam C.I. Oancea B. Ochir C. Odukoya O.O. Ogbo F.A. Olagunju A.T. Olakunde B.O. Omar E. Bali O.A. Omonisi A.E.E. Ong S. Onwujekwe O.E. Orru H. Altamirano O.D.V. Otstavnov N. Otstavnov S.S. Owolabi M.O. /surname> P.M.
  53. Markou A. Chiamulera C. Geyer M.A. Tricklebank M. Steckler T. Removing obstacles in neuroscience drug discovery: The future path for animal models. Neuropsychopharmacology 2009 34 1 74 89 10.1038/npp.2008.173 18830240
    [Google Scholar]
  54. Umbricht D Niggli M Ducray S.P Deptula D Moore R Grünbauer W Boak L Fontoura P. Randomized, double-blind, placebo-controlled trial of the mGlu2/3 negative allosteric modulator decoglurant in partially refractory major depressive disorder. J. Clin. Psych. 2020 81 4 467
    [Google Scholar]
  55. Engers J.L. Bollinger K.A. Weiner R.L. Rodriguez A.L. Long M.F. Breiner M.M. Chang S. Bollinger S.R. Bubser M. Jones C.K. Morrison R.D. Bridges T.M. Blobaum A.L. Niswender C.M. Conn P.J. Emmitte K.A. Lindsley C.W. Design and synthesis of N-aryl phenoxyethoxy pyridinones as highly selective and CNS penetrant mGlu3 NAMs. ACS Med. Chem. Lett. 2017 8 9 925 930 10.1021/acsmedchemlett.7b00249 28947938
    [Google Scholar]
  56. Goeldner C. Ballard T.M. Knoflach F. Wichmann J. Gatti S. Umbricht D. Cognitive impairment in major depression and the mGlu2 receptor as a therapeutic target. Neuropharmacology 2013 64 337 346 10.1016/j.neuropharm.2012.08.001 22992331
    [Google Scholar]
  57. Hascup E.R. Hascup K.N. Stephens M. Pomerleau F. Huettl P. Gratton A. Gerhardt G.A. Rapid microelectrode measurements and the origin and regulation of extracellular glutamate in rat prefrontal cortex. J. Neurochem. 2010 115 6 1608 1620 10.1111/j.1471‑4159.2010.07066.x 20969570
    [Google Scholar]
  58. Khajavi D. Farokhnia M. Modabbernia A. Ashrafi M. Abbasi S.H. Tabrizi M. Akhondzadeh S. Oral scopolamine augmentation in moderate to severe major depressive disorder: A randomized, double-blind, placebo-controlled study. J. Clin. Psychiatry 2012 73 11 1428 1433 10.4088/JCP.12m07706 23146150
    [Google Scholar]
  59. Singh K. Pal R. Khan S.A. Kumar B. Akhtar M.J. Insights into the structure activity relationship of nitrogen-containing heterocyclics for the development of antidepressant compounds: An updated review. J. Mol. Struct. 2021 1237 130369 10.1016/j.molstruc.2021.130369
    [Google Scholar]
  60. Sirakanyan S.N. Spinelli D. Geronikaki A. Kartsev V. Hakobyan E.K. Petrou A. Paronikyan R.G. Nazaryan I.M. Akopyan H.H. Hovakimyan A.A. Synthesis and Neurotropic Activity of New Heterocyclic Systems: Pyridofuro[3,2-d]pyrrolo[1,2-a]pyrimidines, Pyridofuro[3,2-d]pyrido[1,2-a]pyrimidines and Pyridofuro[3′,2′: 4,5]pyrimido[1,2-a]azepines. Molecules 2021 26 11 3320 10.3390/molecules26113320 34205930
    [Google Scholar]
  61. Furukawa T. McGuire H. Barbui C. Low dosage tricyclic antidepressants for depression. Cochrane Database Syst. Rev. 2003 2003 3 CD003197 12917952
    [Google Scholar]
  62. Muñoz L.F. D’Ocón P. Romero A. Guerra J.A. Álamo C. Role of serendipity in the discovery of classical antidepressant drugs: Applying operational criteria and patterns of discovery. World J. Psychiatry 2022 12 4 588 602 10.5498/wjp.v12.i4.588 35582332
    [Google Scholar]
  63. Mu W. Xu G. Wang Z. Li Q. Sun S. Qin Q. Li Z. Shi W. Dai W. Zhan X. Wang J. Bai Z. Xiao X. Tricyclic antidepressants induce liver inflammation by targeting NLRP3 inflammasome activation. Cell Commun. Signal. 2023 21 1 123 10.1186/s12964‑023‑01128‑x 37231437
    [Google Scholar]
  64. Laxmikeshav K. Kumari P. Shankaraiah N. Expedition of sulfur-containing heterocyclic derivatives as cytotoxic agents in medicinal chemistry: A decade update. Med. Res. Rev. 2022 42 1 513 575 10.1002/med.21852 34453452
    [Google Scholar]
  65. Banik BK Sahoo BM Kumar BR Panda J Kumar A Green chemistry and synthetic approaches in the development of antidepressant and antipsychotic agents. Green approaches in medicinal chemistry for sustainable drug design. Elsevier 2024 513 530 10.1016/B978‑0‑12‑817592‑7.00020‑4
    [Google Scholar]
  66. Elmegeed G.A. Baiuomy A.R. Abdelhalim M.M. Hana H.Y. Synthesis and antidepressant evaluation of five novel heterocyclic tryptophan-hybrid derivatives. Arch. Pharm. 2010 343 5 261 267 10.1002/ardp.200900244 20232373
    [Google Scholar]
  67. Bawa S. Siddiqui N. Andalip Ali R. Afzal O. Akhtar M.J. Azad B. Kumar R. Antidepressant potential of nitrogen-containing heterocyclic moieties: An updated review. J. Pharm. Bioallied Sci. 2011 3 2 194 212 10.4103/0975‑7406.80765 21687347
    [Google Scholar]
  68. Carboni L. Rullo L. Caputi F.F. Stamatakos S. Candeletti S. Romualdi P. Chronic trazodone and citalopram treatments increase trophic factor and circadian rhythm gene expression in rat brain regions relevant for antidepressant efficacy. Int. J. Mol. Sci. 2022 23 22 14041 10.3390/ijms232214041 36430520
    [Google Scholar]
  69. Goracci A. Forgione R.N. Giorgi D.R. Coluccia A. Cuomo A. Fagiolini A. Practical guidance for prescribing trazodone extended-release in major depression. Expert Opin. Pharmacother. 2016 17 3 433 441 10.1517/14656566.2016.1133587 26678742
    [Google Scholar]
  70. Cipriani A. Furukawa T.A. Salanti G. Chaimani A. Atkinson L.Z. Ogawa Y. Leucht S. Ruhe H.G. Turner E.H. Higgins J.P.T. Egger M. Takeshima N. Hayasaka Y. Imai H. Shinohara K. Tajika A. Ioannidis J.P.A. Geddes J.R. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: A systematic review and network meta-analysis. Lancet 2018 391 10128 1357 1366 10.1016/S0140‑6736(17)32802‑7 29477251
    [Google Scholar]
  71. harbi A.K.S. Treatment-resistant depression: Therapeutic trends, challenges, and future directions. Patient Prefer. Adherence 2012 6 369 388 10.2147/PPA.S29716 22654508
    [Google Scholar]
  72. Peselow E.D. Filippi A.M. Goodnick P. Barouche F. Fieve R.R. The short- and long-term efficacy of paroxetine HCl: B. Data from a double-blind crossover study and from a year-long term trial vs. imipramine and placebo. Psychopharmacol. Bull. 1989 25 2 272 276 2532374
    [Google Scholar]
  73. Thase M.E. Antidepressant combinations: Cutting edge psychopharmacology or passing fad? Curr. Psychiatry Rep. 2013 15 10 403 10.1007/s11920‑013‑0403‑2 24052267
    [Google Scholar]
  74. Moret C. Combination/augmentation strategies for improving the treatment of depression. Neuropsychiatr. Dis. Treat. 2005 1 4 301 309 18568111
    [Google Scholar]
  75. Nelson J.C. Mazure C.M. Bowers M.B. Jr Jatlow P.I. A preliminary, open study of the combination of fluoxetine and desipramine for rapid treatment of major depression. Arch. Gen. Psychiatry 1991 48 4 303 307 10.1001/archpsyc.1991.01810280019002 2009031
    [Google Scholar]
  76. Holland J.C. Romano S.J. Heiligenstein J.H. Tepner R.G. Wilson M.G. A controlled trial of fluoxetine and desipramine in depressed women with advanced cancer. Psycho-Oncology: Journal of the Psychological. Soc. Behav. Dimen. Cancer. 1998 7 4 291 300
    [Google Scholar]
  77. Egan B.M. Zhao Y. Axon R.N. Brzezinski W.A. Ferdinand K.C. Uncontrolled and apparent treatment resistant hypertension in the United States, 1988 to 2008. Circulation 2011 124 9 1046 1058 10.1161/CIRCULATIONAHA.111.030189 21824920
    [Google Scholar]
  78. Thase M.E. Rush A.J. Kasper S. Nemeroff C.B. Tricyclics and newer antidepressant medications: Treatment options for treatment-resistant depressions. Depression 1994 2 3 152 168 10.1002/depr.3050020307
    [Google Scholar]
  79. Gillman P.K. Tricyclic antidepressant pharmacology and therapeutic drug interactions updated. Br. J. Pharmacol. 2007 151 6 737 748 10.1038/sj.bjp.0707253 17471183
    [Google Scholar]
  80. Pinheiro R.R. Duarte B. Cabete J. Trichloroacetic acid (80%) as a chemical debridement method for chronic venous leg ulcers-A pilot study. Int. Wound J. 2018 15 3 438 440 10.1111/iwj.12884 29334174
    [Google Scholar]
  81. Dantzer R. O’Connor J.C. Freund G.G. Johnson R.W. Kelley K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008 9 1 46 56 10.1038/nrn2297 18073775
    [Google Scholar]
  82. Hestad K.A. Engedal K. Whist J.E. Farup P.G. The relationships among tryptophan, kynurenine, indoleamine 2, 3-dioxygenase, depression, and neuropsychological performance. Front. Psychol. 2017 8 1561 10.3389/fpsyg.2017.01561 29046648
    [Google Scholar]
  83. Dantzer R. Role of the kynurenine metabolism pathway in inflammation-induced depression: Preclinical approaches. Curr Top Behav Neurosci 2017 31 117 138 10.1007/7854_2016_6
    [Google Scholar]
  84. Pathak S. Nadar R. Kim S. Liu K. Govindarajulu M. Cook P. Alexander W.C.S. Dhanasekaran M. Moore T. The influence of kynurenine metabolites on neurodegenerative pathologies. Int. J. Mol. Sci. 2024 25 2 853 10.3390/ijms25020853 38255925
    [Google Scholar]
  85. Onaolapo A.Y. Onaolapo O.J. Glutamate and depression: Reflecting a deepening knowledge of the gut and brain effects of a ubiquitous molecule. World J. Psychiatry 2021 11 7 297 315 10.5498/wjp.v11.i7.297 34327123
    [Google Scholar]
  86. Bryleva E.Y. Brundin L. Kynurenine pathway metabolites and suicidality. Neuropharmacology 2017 112 Pt B 324 330 10.1016/j.neuropharm.2016.01.034 26820800
    [Google Scholar]
  87. Flint A.J. Augmentation strategies in geriatric depression. Int. J. Geriatr. Psychiatry 1995 10 2 137 146 10.1002/gps.930100209
    [Google Scholar]
  88. Ľupták M. Michaličková D. Fišar Z. Kitzlerová E. Hroudová J. Novel approaches in schizophrenia-from risk factors and hypotheses to novel drug targets. World J. Psychiatry 2021 11 7 277 296 10.5498/wjp.v11.i7.277 34327122
    [Google Scholar]
  89. Wincewicz D. Braszko J. Validation of brain angiotensin system blockade as a novel drug target in pharmacological treatment of neuropsychiatric disorders. Pharmacopsychiatry 2017 50 6 233 247 10.1055/s‑0043‑112345 28641333
    [Google Scholar]
  90. Farvolden P. Kennedy S.H. Lam R.W. Recent developments in the psychobiology and pharmacotherapy of depression: Optimising existing treatments and novel approaches for the future. Expert Opin. Investig. Drugs 2003 12 1 65 86 10.1517/13543784.12.1.65 12517255
    [Google Scholar]
  91. Jiao S. Cao T. Cai H. Peripheral biomarkers of treatment-resistant schizophrenia: Genetic, inflammation and stress perspectives. Front. Pharmacol. 2022 13 1005702 10.3389/fphar.2022.1005702 36313375
    [Google Scholar]
  92. Stewart A.M. Kalueff A.V. Anxiolytic drug discovery: What are the novel approaches and how can we improve them? Expert Opin. Drug Discov. 2014 9 1 15 26 10.1517/17460441.2014.857309 24206163
    [Google Scholar]
  93. Tang J. Chen L.R. Chen K.H. The utilization of dehydroepiandrosterone as a sexual hormone precursor in premenopausal and postmenopausal women: An overview. Pharmaceuticals 2021 15 1 46 10.3390/ph15010046 35056103
    [Google Scholar]
  94. Labrie F. DHEA, important source of sex steroids in men and even more in women. Prog. Brain Res. 2010 182 97 148 10.1016/S0079‑6123(10)82004‑7 20541662
    [Google Scholar]
  95. Kilanczyk E. Ruminkiewicz D. Banales J.M. Milkiewicz P. Milkiewicz M. DHEA protects human cholangiocytes and hepatocytes against apoptosis and oxidative stress. Cells 2022 11 6 1038 10.3390/cells11061038 35326489
    [Google Scholar]
  96. Misiak B. Piotrowski P. Chęć M. Samochowiec J. Cortisol and dehydroepiandrosterone sulfate in patients with schizophrenia spectrum disorders with respect to cognitive performance. Compr. Psychoneuroendocrinol. 2021 6 100041 10.1016/j.cpnec.2021.100041 35757369
    [Google Scholar]
  97. Gould T.D. Chen G. Manji H.K. Mood stabilizer psychopharmacology. Clin. Neurosci. Res. 2002 2 3-4 193 212 10.1016/S1566‑2772(02)00044‑0 22707923
    [Google Scholar]
  98. Leo R.J. Narendran R. Anticonvulsant use in the treatment of bipolar disorder: A primer for primary care physicians. Prim. Care Companion J. Clin. Psychiatry 1999 1 3 74 84 15014689
    [Google Scholar]
  99. Grunze A. Amann B.L. Grunze H. Efficacy of carbamazepine and its derivatives in the treatment of bipolar disorder. Medicina 2021 57 5 433 10.3390/medicina57050433 33946323
    [Google Scholar]
  100. Ward M.P. Irazoqui P.P. Evolving refractory major depressive disorder diagnostic and treatment paradigms: Toward closed-loop therapeutics. Front. Neuroeng. 2010 3 7 10.3389/fneng.2010.00007 20631824
    [Google Scholar]
  101. Vieta E. Moreno S.J. Acute and long-term treatment of mania. Dialogues Clin. Neurosci. 2008 10 2 165 179 10.31887/DCNS.2008.10.2/evieta 18689287
    [Google Scholar]
  102. Khanna S. Vieta E. Lyons B. Grossman F. Eerdekens M. Kramer M. Risperidone in the treatment of acute mania. Br. J. Psychiatry 2005 187 3 229 234 10.1192/bjp.187.3.229 16135859
    [Google Scholar]
  103. Polese D. Fornaro M. Palermo M. Luca D.V. Bartolomeis d.A. Treatment-resistant to antipsychotics: A resistance to everything? Psychotherapy in treatment-resistant schizophrenia and nonaffective psychosis: A 25-year systematic review and exploratory meta-analysis. Front. Psychiatry 2019 10 210 10.3389/fpsyt.2019.00210 31057434
    [Google Scholar]
  104. Kolovos S. Tulder v.M.W. Cuijpers P. Prigent A. Chevreul K. Riper H. Bosmans J.E. The effect of treatment as usual on major depressive disorder: A meta-analysis. J. Affect. Disord. 2017 210 72 81 10.1016/j.jad.2016.12.013 28013125
    [Google Scholar]
  105. Bonson K. Buckholtz J.W. Murphy D.L. Chronic administration of serotonergic antidepressants attenuates the subjective effects of LSD in humans. Neuropsychopharmacology 1996 14 6 425 436 10.1016/0893‑133X(95)00145‑4 8726753
    [Google Scholar]
  106. Petermann F. Hospital anxiety and depression scale, deutsche version (HADS-D). Z. Psychiatr. Psychol. Psychother. 2015
    [Google Scholar]
  107. Gasser P. Holstein D. Michel Y. Doblin R. Klosinski Y.B. Passie T. Brenneisen R. Safety and efficacy of lysergic acid diethylamide-assisted psychotherapy for anxiety associated with life-threatening diseases. J. Nerv. Ment. Dis. 2014 202 7 513 520 10.1097/NMD.0000000000000113 24594678
    [Google Scholar]
  108. Páleníček T. Hliňák Z. Bubeníková-Valešová V. Novák T. Horáček J. Sex differences in the effects of N,N-diethyllysergamide (LSD) on behavioural activity and prepulse inhibition. Prog. Neuropsychopharmacol. Biol. Psychiatry 2010 34 4 588 596 10.1016/j.pnpbp.2010.02.008 20156516
    [Google Scholar]
  109. Thiessen M.S. Walsh Z. Bird B.M. Lafrance A. Psychedelic use and intimate partner violence: The role of emotion regulation. J. Psychopharmacol. 2018 32 7 749 755 10.1177/0269881118771782 29807492
    [Google Scholar]
  110. Harris C.R.L. Bolstridge M. Day C.M.J. Rucker J. Watts R. Erritzoe D.E. Kaelen M. Giribaldi B. Bloomfield M. Pilling S. Rickard J.A. Forbes B. Feilding A. Taylor D. Curran H.V. Nutt D.J. Psilocybin with psychological support for treatment-resistant depression: Six-month follow-up. Psychopharmacology 2018 235 2 399 408 10.1007/s00213‑017‑4771‑x 29119217
    [Google Scholar]
  111. Roseman L. Nutt D.J. Harris C.R.L. Quality of acute psychedelic experience predicts therapeutic efficacy of psilocybin for treatment-resistant depression. Front. Pharmacol. 2018 8 974 10.3389/fphar.2017.00974 29387009
    [Google Scholar]
  112. Liebes A.G. The role of self-compassion in psilocybin-assisted motivational enhancement therapy to treat alcohol dependence: A randomized controlled trial. Psychol. Addict. Behav. 2019 38 1 101 113
    [Google Scholar]
  113. Gerhard D.M. Pothula S. Liu R.J. Wu M. Li X.Y. Girgenti M.J. Taylor S.R. Duman C.H. Delpire E. Picciotto M. Wohleb E.S. Duman R.S. GABA interneurons are the cellular trigger for ketamine’s rapid antidepressant actions. J. Clin. Invest. 2020 130 3 1336 1349 10.1172/JCI130808 31743111
    [Google Scholar]
  114. Kavalali E.T. Monteggia L.M. Targeting homeostatic synaptic plasticity for treatment of mood disorders. Neuron 2020 106 5 715 726 10.1016/j.neuron.2020.05.015 32497508
    [Google Scholar]
  115. Holmes S.E. Gallezot J.D. Davis M.T. DellaGioia N. Matuskey D. Nabulsi N. Krystal J.H. Javitch J.A. DeLorenzo C. Carson R.E. Esterlis I. Measuring the effects of ketamine on mGluR5 using [ 18 F]FPEB and PET. J. Cereb. Blood Flow Metab. 2020 40 11 2254 2264 10.1177/0271678X19886316 31744389
    [Google Scholar]
  116. Kato T. Pothula S. Liu R.J. Duman C.H. Terwilliger R. Vlasuk G.P. Saiah E. Hahm S. Duman R.S. Sestrin modulator NV-5138 produces rapid antidepressant effects via direct mTORC1 activation. J. Clin. Invest. 2019 129 6 2542 2554 10.1172/JCI126859 30990795
    [Google Scholar]
  117. Nugent A.C. Robinson S.E. Coppola R. Zarate C.A. Jr Preliminary differences in resting state MEG functional connectivity pre- and post-ketamine in major depressive disorder. Psychiatry Res. Neuroimaging 2016 254 56 66 10.1016/j.pscychresns.2016.06.006 27362845
    [Google Scholar]
  118. Abdallah C.G. Averill L.A. Collins K.A. Geha P. Schwartz J. Averill C. DeWilde K.E. Wong E. Anticevic A. Tang C.Y. Iosifescu D.V. Charney D.S. Murrough J.W. Ketamine treatment and global brain connectivity in major depression. Neuropsychopharmacology 2017 42 6 1210 1219 10.1038/npp.2016.186 27604566
    [Google Scholar]
  119. Tedesco S. Gajaram G. Chida S. Ahmad A. Pentak M. Kelada M. Lewis L. Krishnan D. Tran C. Soetan O.T. Mukona L.T. Jolayemi A. The efficacy of MDMA (3, 4-Methylenedioxymethamphetamine) for post-traumatic stress disorder in humans: A systematic review and meta-analysis. Cureus 2021 13 5 e15070 10.7759/cureus.15070 34150406
    [Google Scholar]
  120. Riaz K. Suneel S. Hamza Bin Abdul Malik M. Kashif T. Ullah I. Waris A. Nicola D.M. Mazza M. Sani G. Martinotti G. Berardis D.D. MDMA-based psychotherapy in treatment-resistant post-traumatic stress disorder (PTSD): A brief narrative overview of current evidence. Diseases 2023 11 4 159 10.3390/diseases11040159 37987270
    [Google Scholar]
  121. García M.J.A. de la Fuente Revenga M. Gil A.S. Franco R.M.I. Feilding A. Castillo P.A. Riba J. The alkaloids of Banisteriopsis caapi, the plant source of the Amazonian hallucinogen Ayahuasca, stimulate adult neurogenesis in vitro. Sci. Rep. 2017 7 1 5309 10.1038/s41598‑017‑05407‑9 28706205
    [Google Scholar]
  122. Osório F.L. Sanches R.F. Macedo L.R. Santos d.R.G. Maia-de-Oliveira J.P. Ana W.L. Araujo d.D.B. Riba J. Crippa J.A. Hallak J.E. Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: A preliminary report. Rev. Bras. Psiquiatr. 2015 37 1 13 20 10.1590/1516‑4446‑2014‑1496 25806551
    [Google Scholar]
  123. Ragnhildstveit A. Khan R. Seli P. Bass L.C. August R.J. Kaiyo M. Barr N. Jackson L.K. Gaffrey M.S. Barsuglia J.P. Averill L.A. 5-MeO-DMT for post-traumatic stress disorder: A real-world longitudinal case study. Front. Psychiatry 2023 14 1271152 10.3389/fpsyt.2023.1271152 38076677
    [Google Scholar]
  124. Reckweg J.T. Uthaug M.V. Szabo A. Davis A.K. Lancelotta R. Mason N.L. Ramaekers J.G. The clinical pharmacology and potential therapeutic applications of 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT). J. Neurochem. 2022 162 1 128 146 10.1111/jnc.15587 35149998
    [Google Scholar]
  125. Ermakova A.O. Dunbar F. Rucker J. Johnson M.W. A narrative synthesis of research with 5-MeO-DMT. J. Psychopharmacol. 2022 36 3 273 294 10.1177/02698811211050543 34666554
    [Google Scholar]
  126. Germann C.B. 5-methoxy-N, N-dimethyltryptamine: An ego-dissolving endogenous neurochemical catalyst of creativity. Act. Nerv. Super. 2019 61 4 170 216 10.1007/s41470‑019‑00063‑y
    [Google Scholar]
  127. Rush B Marcus O Shore R Cunningham L Thompson N Rideout K. Psychedelic medicine: A rapid review of therapeutic applications and implications for future research. Homewood Research Institute Guelph, ON, Canada 2022
    [Google Scholar]
  128. Dawood Hristova J.J. Pérez-Jover V. Psychotherapy with psilocybin for depression: systematic review. Behav. Sci. (Basel) 2023 13 4 297
    [Google Scholar]
/content/journals/cmc/10.2174/0109298673342524250109181220
Loading
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test