Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects

G Sanacora, M A Smith, S Pathak, H-L Su, P H Boeijinga, D J McCarthy, M C Quirk, G Sanacora, M A Smith, S Pathak, H-L Su, P H Boeijinga, D J McCarthy, M C Quirk

Abstract

Ketamine, an N-methyl-D-aspartate receptor (NMDAR) channel blocker, has been found to induce rapid and robust antidepressant-like effects in rodent models and in treatment-refractory depressed patients. However, the marked acute psychological side effects of ketamine complicate the interpretation of both preclinical and clinical data. Moreover, the lack of controlled data demonstrating the ability of ketamine to sustain the antidepressant response with repeated administration leaves the potential clinical utility of this class of drugs in question. Using quantitative electroencephalography (qEEG) to objectively align doses of a low-trapping NMDA channel blocker, AZD6765 (lanicemine), to that of ketamine, we demonstrate the potential for NMDA channel blockers to produce antidepressant efficacy without psychotomimetic and dissociative side effects. Furthermore, using placebo-controlled data, we show that the antidepressant response to NMDA channel blockers can be maintained with repeated and intermittent drug administration. Together, these data provide a path for the development of novel glutamatergic-based therapeutics for treatment-refractory mood disorders.

Trial registration: ClinicalTrials.gov NCT00491686 NCT00781742 NCT01130909.

Figures

Figure 1
Figure 1
Electroencephalography (EEG) effects of lanicemine relative to ketamine in rodent model. Left: Time course of gamma-band EEG following administration of lanicemine and ketamine (doses: 10 mg kg−1) and respective vehicles. Right: Tolerability of lanicemine vs ketamine, measured by hyper-locomotor activity, at comparable levels of target engagement.
Figure 2
Figure 2
Electroencephalography (EEG) effects of lanicemine relative to ketamine in man. Left: Distribution over skull surface of absolute gamma EEG magnitude, theta-cordance and alpha slow-wave index at 1 h of perfusion. Color mapping depicts levels of statistical significance between study drug (lanicemine 75 or 150 mg; ketamine 0.5 mg kg−1) and vehicle at a given electrode location (blue: decrease; red: increase in P-value). Top right: Comparison of lanicemine (75 or 150 mg), ketamine (0.5 mg kg−1), and vehicle for mean (s.e.) change in relative gamma-EEG magnitude over time (eyes closed). Bottom right: Comparison of lanicemine (75 or 150 mg), ketamine (0.5 mg kg−1), and vehicle for mean (s.e.) change in Clinician Administered Dissociative States Scale (CADSS) total score from baseline. CADSS total score was increased significantly with ketamine vs vehicle at 1 h (P<0.01) and across all times (P<0.05). Lanicemine (each dose) did not significantly increase CADSS total score vs vehicle at any time.
Figure 3
Figure 3
Montgomery Åsberg Depression rating Scale (MADRS) score change at prespecified time points during the 3-week treatment and 5-week follow-up period in lanicemine 100 mg, lanicemine 150 mg and placebo groups (intent-to-treat (ITT), last observation carried forward (LOCF)) (phase IIB study, study 9).
Figure 4
Figure 4
Clinical Global Impression of Improvement (CGI-I) response rate (%) at prespecified time points during the 3-week treatment and 5-week follow-up period in lanicemine 100 mg, lanicemine 150 mg and placebo groups. Response is defined as CGI-I score⩽2 (much or very much improved) (phase IIB study, study 9).

References

    1. World Health Organization The Global Burden of Disease: 2004 Update WHO Press: Geneva, Switzerland; Available at. Accessed 14 September2012
    1. Insel TR, Wang PS. The STAR*D trial: revealing the need for better treatments. Psychiatr Serv. 2009;60:1466–1467.
    1. Popoli M, Yan Z, McEwen BS, Sanacora G. The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci. 2012;13:22–37.
    1. 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.
    1. Sanacora G, Zarate CA, Krystal JH, Manji HK. Targeting the glutamatergic system to develop novel, improved therapeutics for mood disorders. Nat Rev Drug Discov. 2008;7:426–437.
    1. Aan Het RM, Zarate CA, Jr., Charney DS, Mathew SJ. Ketamine for depression: where do we go from here. Biol Psychiatry. 2012;72:537–547.
    1. 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.
    1. Maeng S, Zarate CA, Jr., Du J, Schloesser RJ, McCammon J, Chen G, et al. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol Psychiatry. 2008;63:349–352.
    1. Quirk MC, Sosulski DL, Feierstein CE, Uchida N, Mainen ZF. A defined network of fast-spiking interneurons in orbitofrontal cortex: responses to behavioral contingencies and ketamine administration. Front Syst Neurosci. 2009;3:13.
    1. Homayoun H, Moghaddam B. NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci. 2007;27:11496–11500.
    1. Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012;338:68–72.
    1. 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.
    1. Coyle JT. The GABA-glutamate connection in schizophrenia: which is the proximate cause. Biochem Pharmacol. 2004;68:1507–1514.
    1. Mealing GA, Lanthorn TH, Murray CL, Small DL, Morley P. Differences in degree of trapping of low-affinity uncompetitive N-methyl-D-aspartic acid receptor antagonists with similar kinetics of block. J Pharmacol Exp Ther. 1999;288:204–210.
    1. Mealing GA, Lanthorn TH, Small DL, Murray RJ, Mattes KC, Comas TM, et al. Structural modifications to an N-methyl-D-aspartate receptor antagonist result in large differences in trapping block. J Pharmacol Exp Ther. 2001;297:906–914.
    1. Matejcek M. Cortical correlates of vigilance regulation and their use in evaluating the effects of treatment. Adv Biochem Psychopharmacol. 1980;23:339–348.
    1. Jobert M, Wilson FJ, Ruigt GS, Brunovsky M, Prichep LS, Drinkenburg WH. Guidelines for the recording and evaluation of pharmaco-EEG data in man: the International Pharmaco-EEG Society (IPEG) Neuropsychobiology. 2012;66:201–220.
    1. Rush AJ, Warden D, Wisniewski SR, Fava M, Trivedi MH, Gaynes BN, et al. STAR*D: revising conventional wisdom. CNS Drugs. 2009;23:627–647.
    1. Balestra M, Bernstein P, Ernst GE, Frietze W, McCauley JP, Nugiel D, et al. (inventors). AstraZeneca Pharmaceuticals LP (assignee). Ethanamine Compounds and Methods of Using the Same. Patent US20120277272 A12012
    1. Dravid SM, Erreger K, Yuan H, Nicholson K, Le P, Lyuboslavsky P, et al. Subunit-specific mechanisms and proton sensitivity of NMDA receptor channel block. J Physiol. 2007;581:107–128.
    1. Foster AC, Wong EH. The novel anticonvulsant MK-801 binds to the activated state of the N-methyl-D-aspartate receptor in rat brain. Br J Pharmacol. 1987;91:403–409.
    1. Gilling KE, Jatzke C, Hechenberger M, Parsons CG. Potency, voltage-dependency, agonist concentration-dependency, blocking kinetics and partial untrapping of the uncompetitive N-methyl-D-aspartate (NMDA) channel blocker memantine at human NMDA (GluN1/GluN2A) receptors. Neuropharmacology. 2009;56:866–875.
    1. Hakami T, Jones NC, Tolmacheva EA, Gaudias J, Chaumont J, Salzberg M, et al. NMDA receptor hypofunction leads to generalized and persistent aberrant gamma oscillations independent of hyperlocomotion and the state of consciousness. PLoS ONE. 2009;4:e6755.
    1. Kocsis B. Differential role of NR2A and NR2B subunits in N-methyl-D-aspartate receptor antagonist-induced aberrant cortical gamma oscillations. Biol Psychiatry. 2012;71:987–995.
    1. Cook IA, Leuchter AF, Morgan M, Witte E, Stubbeman WF, Abrams M, et al. Early changes in prefrontal activity characterize clinical responders to antidepressants. Neuropsychopharmacology. 2002;27:120–131.
    1. Horacek J, Brunovsky M, Novak T, Tislerova B, Palenicek T, Bubenikova-Valesova V, et al. Subanesthetic dose of ketamine decreases prefrontal theta cordance in healthy volunteers: implications for antidepressant effect. Psychol Med. 2010;40:1443–1451.
    1. Boeijinga PH, Parot P, Soufflet L, Landron F, Danel T, Gendre I, et al. Pharmacodynamic effects of acamprosate on markers of cerebral function in alcohol-dependent subjects administered as pretreatment and during alcohol abstinence. Neuropsychobiology. 2004;50:71–77.
    1. Palmer GC, Miller JA, Cregan EF, Gendron P, Peeling J. Low-affinity NMDA receptor antagonists. The neuroprotective potential of ARL 15896AR. Ann NY Acad Sci. 1997;825:220–231.
    1. 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.
    1. Zarate CA, Jr., Mathews D, Ibrahim L, Chaves JF, Marquardt C, Ukoh I, et al. Replication of ketamine's antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry. 2012;71:939–946.
    1. Murrough JW, Perez AM, Pillemer S, Stern J, Parides MK, Aan Het RM, et al. Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry. 2012;74:250–256.
    1. Burgdorf J, Zhang XL, Nicholson KL, Balster RL, Leander JD, Stanton PK, et al. GLYX-13, a NMDA receptor glycine-site functional partial agonist, induces antidepressant-like effects without ketamine-like side effects. Neuropsychopharmacology. 2013;38:729–742.
    1. Chowdhury GM, Behar KL, Cho W, Thomas MA, Rothman DL, Sanacora G. 1H-[13C]-nuclear magnetic resonance spectroscopy measures of ketamine's effect on amino acid neurotransmitter metabolism. Biol Psychiatry. 2012;71:1022–1025.
    1. Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17:2921–2927.
    1. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011;475:91–95.
    1. Zarate CA, Jr., Mathews D, Ibrahim L, Chaves JF, Marquardt C, Ukoh I, et al. A randomized trial of a low-trapping nonselective N-Methyl-D-Aspartate channel blocker in major depression. Biol Psychiatry. 2012;74:257–264.
    1. Cusin C, Soskin DP, Kara PJ, Kelley D, Trina C, Paolo C, et al. Open-label, flexible-dose repeated intravenous ketamine infusions as adjunct in outpatients with treatment resistant major depression with suicidal ideation New Research Approaches for Mental Health Interactions28–31 May2013. Abstract 43.

Source: PubMed

3
订阅