Other N-Methyl-D-Aspartate (NMDA) Receptor Antagonists with a Rapid Onset of Action and Less Side Effect in the Treatment of Depression

우울증 치료에서 빠른 효과와 적은 부작용을 가진 새로운 N-Methyl-D-Aspartate(NMDA) 수용체 길항제

  • Choi, Bum-Sung (Department of Psychiatry, Dankook University Cheonan Hospital, Dankook University College of Medicine) ;
  • Lee, Hwa-Young (Department of Psychiatry, Soonchunhyang Hospital Cheonan, Soonchunhyang University College of Medicine)
  • 최범성 (단국대학교 의과대학 천안병원 정신건강의학교실) ;
  • 이화영 (순천향대학교 의과대학 천안병원 정신건강의학교실)
  • Received : 2015.09.25
  • Accepted : 2015.11.03
  • Published : 2015.11.30

Abstract

Mood disorder is a common psychiatric illness with a high lifetime prevalence in the general population. Many prescribed antidepressants modulate monoamine neurotransmitters including serotonin, norepinephrine and dopamine. There has been greater focus on the major excitatory neurotransmitter in the human brain, glutamate, in the pathophysiology and treatment of major depressive disorder (MDD). Recently, ketamine, an N-methyl-D-aspartate receptor antagonist, has received attention and has been investigated for clinical trials and neurobiological studies. In this article, we will review the clinical evidence for glutamatergic dysfunction in MDD, the progress with ketamine as a rapidly acting antidepressant, and other N-methyl-D-aspartate receptor antagonist for treatment-resistant depression.

Keywords

References

  1. Kessler RC, McGonagle KA, Zhao S, Nelson CB, Hughes M, Eshleman S, et al. Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbidity Survey. Arch Gen Psychiatry 1994;51:8-19. https://doi.org/10.1001/archpsyc.1994.03950010008002
  2. Montgomery SA, Bebbington P, Cowen P, Deakin W, Freeling P, Hallstrom C, et al. Guidelines for treating depressive illness with antidepressants: a statement from the British Association for Psychopharmacology. J Psychopharmacol 1993;7(1 Suppl):19-23. https://doi.org/10.1177/0269881193007001041
  3. Hirschfeld RM, Keller MB, Panico S, Arons BS, Barlow D, Davidoff F, et al. The National Depressive and Manic-Depressive Association consensus statement on the undertreatment of depression. JAMA 1997;277:333-340. https://doi.org/10.1001/jama.1997.03540280071036
  4. Massart R, Mongeau R, Lanfumey L. Beyond the monoaminergic hypothesis: neuroplasticity and epigenetic changes in a transgenic mouse model of depression. Philos Trans R Soc Lond B Biol Sci 2012;367:2485-2494. https://doi.org/10.1098/rstb.2012.0212
  5. Rajkowska G. Postmortem studies in mood disorders indicate altered numbers of neurons and glial cells. Biol Psychiatry 2000;48:766-777. https://doi.org/10.1016/S0006-3223(00)00950-1
  6. Smialowska M, Szewczyk B, Wozniak M, Wawrzak-Wlecial A, Domin H. Glial degeneration as a model of depression. Pharmacol Rep 2013;65:1572-1579. https://doi.org/10.1016/S1734-1140(13)71518-4
  7. Hashimoto K, Sawa A, Iyo M. Increased levels of glutamate in brains from patients with mood disorders. Biol Psychiatry 2007;62:1310-1316. https://doi.org/10.1016/j.biopsych.2007.03.017
  8. Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B. Reduced levels of NR2A and NR2B subunits of NMDA receptor and PSD- 95 in the prefrontal cortex in major depression. Prog Neuropsychopharmacol Biol Psychiatry 2009;33:70-75. https://doi.org/10.1016/j.pnpbp.2008.10.005
  9. Mayer ML, Westbrook GL. The physiology of excitatory amino acids in the vertebrate central nervous system. Prog Neurobiol 1987;28:197-276. https://doi.org/10.1016/0301-0082(87)90011-6
  10. Chourbaji S, Vogt MA, Fumagalli F, Sohr R, Frasca A, Brandwein C, et al. AMPA receptor subunit 1 (GluR-A) knockout mice model the glutamate hypothesis of depression. FASEB J 2008;22:3129-3134. https://doi.org/10.1096/fj.08-106450
  11. Chang EH, Savage MJ, Flood DG, Thomas JM, Levy RB, Mahadomrongkul V, et al. AMPA receptor downscaling at the onset of Alzheimer’s disease pathology in double knockin mice. Proc Natl Acad Sci U S A 2006;103:3410-3415. https://doi.org/10.1073/pnas.0507313103
  12. Yang J, Shen J. In vivo evidence for reduced cortical glutamate-glutamine cycling in rats treated with the antidepressant/antipanic drug phenelzine. Neuroscience 2005;135:927-937. https://doi.org/10.1016/j.neuroscience.2005.06.067
  13. Paul IA, Skolnick P. Glutamate and depression: clinical and preclinical studies. Ann N Y Acad Sci 2003;1003:250-272. https://doi.org/10.1196/annals.1300.016
  14. Skolnick P, Layer RT, Popik P, Nowak G, Paul IA, Trullas R. Adaptation of N-methyl-D-aspartate (NMDA) receptors following antidepressant treatment: implications for the pharmacotherapy of depression. Pharmacopsychiatry 1996;29:23-26. https://doi.org/10.1055/s-2007-979537
  15. Collingridge GL, Olsen RW, Peters J, Spedding M. A nomenclature for ligand-gated ion channels. Neuropharmacology 2009;56:2-5. https://doi.org/10.1016/j.neuropharm.2008.06.063
  16. Bredt DS, Nicoll RA. AMPA receptor trafficking at excitatory synapses. Neuron 2003;40:361-379. https://doi.org/10.1016/S0896-6273(03)00640-8
  17. Malenka RC, Bear MF. LTP and LTD: an embarrassment of riches. Neuron 2004;44:5-21. https://doi.org/10.1016/j.neuron.2004.09.012
  18. Huettner JE. Kainate receptors and synaptic transmission. Prog Neurobiol 2003;70:387-407. https://doi.org/10.1016/S0301-0082(03)00122-9
  19. Boyer PA, Skolnick P, Fossom LH. Chronic administration of imipramine and citalopram alters the expression of NMDA receptor subunit mRNAs in mouse brain. A quantitative in situ hybridization study. J Mol Neurosci 1998;10:219-233. https://doi.org/10.1007/BF02761776
  20. Sanacora G, Rothman DL, Mason G, Krystal JH. Clinical studies implementing glutamate neurotransmission in mood disorders. Ann N Y Acad Sci 2003;1003:292-308. https://doi.org/10.1196/annals.1300.018
  21. Skolnick P, Legutko B, Li X, Bymaster FP. Current perspectives on the development of non-biogenic amine-based antidepressants. Pharmacol Res 2001;43:411-423. https://doi.org/10.1006/phrs.2000.0806
  22. Zarate CA Jr, Du J, Quiroz J, Gray NA, Denicoff KD, Singh J, et al. Regulation of cellular plasticity cascades in the pathophysiology and treatment of mood disorders: role of the glutamatergic system. Ann N Y Acad Sci 2003;1003:273-291. https://doi.org/10.1196/annals.1300.017
  23. Erecińska M, Silver IA. Metabolism and role of glutamate in mammalian brain. Prog Neurobiol 1990;35:245-296. https://doi.org/10.1016/0301-0082(90)90013-7
  24. Manji HK, Gottesman II, Gould TD. Signal transduction and genesto-behaviors pathways in psychiatric diseases. Sci STKE 2003;2003:pe49.
  25. Bonanno G, Giambelli R, Raiteri L, Tiraboschi E, Zappettini S, Musazzi L, et al. Chronic antidepressants reduce depolarizationevoked glutamate release and protein interactions favoring formation of SNARE complex in hippocampus. J Neurosci 2005;25:3270-3279. https://doi.org/10.1523/JNEUROSCI.5033-04.2005
  26. Magarinos AM, Deslandes A, McEwen BS. Effects of antidepressants and benzodiazepine treatments on the dendritic structure of CA3 pyramidal neurons after chronic stress. Eur J Pharmacol 1999;371:113-122. https://doi.org/10.1016/S0014-2999(99)00163-6
  27. Magarinos AM, McEwen BS. Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: involvement of glucocorticoid secretion and excitatory amino acid receptors. Neuroscience 1995;69:89-98. https://doi.org/10.1016/0306-4522(95)00259-L
  28. Reznikov LR, Grillo CA, Piroli GG, Pasumarthi RK, Reagan LP, Fadel J. Acute stress-mediated increases in extracellular glutamate levels in the rat amygdala: differential effects of antidepressant treatment. Eur J Neurosci 2007;25:3109-3114. https://doi.org/10.1111/j.1460-9568.2007.05560.x
  29. Lowy MT, Gault L, Yamamoto BK. Adrenalectomy attenuates stressinduced elevations in extracellular glutamate concentrations in the hippocampus. J Neurochem 1993;61:1957-1960. https://doi.org/10.1111/j.1471-4159.1993.tb09839.x
  30. Lowy MT, Wittenberg L, Yamamoto BK. Effect of acute stress on hippocampal glutamate levels and spectrin proteolysis in young and aged rats. J Neurochem 1995;65:268-274.
  31. Mark LP, Prost RW, Ulmer JL, Smith MM, Daniels DL, Strottmann JM, et al. Pictorial review of glutamate excitotoxicity: fundamental concepts for neuroimaging. AJNR Am J Neuroradiol 2001;22:1813-1824.
  32. Cameron HA, McEwen BS, Gould E. Regulation of adult neurogenesis by excitatory input and NMDA receptor activation in the dentate gyrus. J Neurosci 1995;15:4687-4692. https://doi.org/10.1523/JNEUROSCI.15-06-04687.1995
  33. Auer DP, Putz B, Kraft E, Lipinski B, Schill J, Holsboer F. Reduced glutamate in the anterior cingulate cortex in depression: an in vivo proton magnetic resonance spectroscopy study. Biol Psychiatry 2000; 47:305-313. https://doi.org/10.1016/S0006-3223(99)00159-6
  34. Schmid RL, Sandler AN, Katz J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain 1999;82:111-125. https://doi.org/10.1016/S0304-3959(99)00044-5
  35. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 1994;51:199-214. https://doi.org/10.1001/archpsyc.1994.03950030035004
  36. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000;47:351-354. https://doi.org/10.1016/S0006-3223(99)00230-9
  37. Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 2006;63:856-864. https://doi.org/10.1001/archpsyc.63.8.856
  38. Zarate CA Jr, Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A, et al. Replication of ketamine's antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry 2012;71:939-946. https://doi.org/10.1016/j.biopsych.2011.12.010
  39. Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, et al. A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry 2010;67:793-802. https://doi.org/10.1001/archgenpsychiatry.2010.90
  40. Duman RS, Li N, Liu RJ, Duric V, Aghajanian G. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology 2012;62:35-41. https://doi.org/10.1016/j.neuropharm.2011.08.044
  41. aan het Rot M, Collins KA, Murrough JW, Perez AM, Reich DL, Charney DS, et al. Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry 2010;67:139-145. https://doi.org/10.1016/j.biopsych.2009.08.038
  42. Reus GZ, Stringari RB, Kirsch TR, Fries GR, Kapczinski F, Roesler R, et al. Neurochemical and behavioural effects of acute and chronic memantine administration in rats: Further support for NMDA as a new pharmacological target for the treatment of depression? Brain Res Bull 2010;81:585-589. https://doi.org/10.1016/j.brainresbull.2009.11.013
  43. Quan MN, Zhang N, Wang YY, Zhang T, Yang Z. Possible antidepressant effects and mechanisms of memantine in behaviors and synaptic plasticity of a depression rat model. Neuroscience 2011;182:88-97. https://doi.org/10.1016/j.neuroscience.2011.03.026
  44. Zarate CA Jr, Singh JB, Quiroz JA, De Jesus G, Denicoff KK, Luckenbaugh DA, et al. A double-blind, placebo-controlled study of memantine in the treatment of major depression. Am J Psychiatry 2006;163:153-155. https://doi.org/10.1176/appi.ajp.163.1.153
  45. Anand A, Gunn AD, Barkay G, Karne HS, Nurnberger JI, Mathew SJ, et al. Early antidepressant effect of memantine during augmentation of lamotrigine inadequate response in bipolar depression: a double-blind, randomized, placebo-controlled trial. Bipolar Disord 2012;14:64-70. https://doi.org/10.1111/j.1399-5618.2011.00971.x
  46. Kollmar R, Markovic K, Thürauf N, Schmitt H, Kornhuber J. Ketamine followed by memantine for the treatment of major depression. Aust N Z J Psychiatry 2008;42:170. https://doi.org/10.1080/00048670701787628
  47. Gideons ES, Kavalali ET, Monteggia LM. Mechanisms underlying differential effectiveness of memantine and ketamine in rapid antidepressant responses. Proc Natl Acad Sci U S A 2014;111:8649-8654. https://doi.org/10.1073/pnas.1323920111
  48. Mealing GA, Lanthorn TH, Murray CL, Small DL, Morley P. Differences in degree of trapping of low-affinity uncompetitive Nmethyl-D-aspartic acid receptor antagonists with similar kinetics of block. J Pharmacol Exp Ther 1999;288:204-210.
  49. Zarate CA Jr, Mathews D, Ibrahim L, Chaves JF, Marquardt C, Ukoh I, et al. A randomized trial of a low-trapping nonselective Nmethyl- D-aspartate channel blocker in major depression. Biol Psychiatry 2013;74:257-264. https://doi.org/10.1016/j.biopsych.2012.10.019
  50. Sanacora G, Smith MA, Pathak S, Su HL, Boeijinga PH, McCarthy DJ, et al. Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects. Mol Psychiatry 2014;19:978-985. https://doi.org/10.1038/mp.2013.130
  51. Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 2010;329:959-964. https://doi.org/10.1126/science.1190287
  52. Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, et al. Glutamate Nmethyl-D-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry 2011;69:754-761. https://doi.org/10.1016/j.biopsych.2010.12.015
  53. Preskorn SH, Baker B, Kolluri S, Menniti FS, Krams M, Landen JW. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol 2008;28:631-637. https://doi.org/10.1097/JCP.0b013e31818a6cea
  54. Ibrahim L, Diaz Granados N, Jolkovsky L, Brutsche N, Luckenbaugh DA, Herring WJ, 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. https://doi.org/10.1097/JCP.0b013e31825d70d6